메뉴 건너뛰기




Volumn 16, Issue 3, 2016, Pages 259-277

Tau-directed approaches for the treatment of Alzheimers disease: Focus on leuco-methylthioninium

Author keywords

Alzheimer's disease; dementia; immunotherapy; leucomethylthioninium; methylthioninium; microtubule stabilizing agents; tau aggregation inhibitors; tau phosphorylation inhibitors; tau protein; TRx0237

Indexed keywords

LEUCO METHYLTHIONINIUM BLUE; METHYLENE BLUE; NOOTROPIC AGENT; TAU PROTEIN; TRX 0237; UNCLASSIFIED DRUG;

EID: 84959325967     PISSN: 14737175     EISSN: 17448360     Source Type: Journal    
DOI: 10.1586/14737175.2016.1140039     Document Type: Review
Times cited : (36)

References (197)
  • 1
    • 84925880097 scopus 로고    scopus 로고
    • 2015 Alzheimers disease facts and figures
    • Alzheimers Association
    • Alzheimers Association. 2015 Alzheimers disease facts and figures. Alzheimers Dement. 2015; 11 (3): 332-384
    • (2015) Alzheimers Dement , vol.11 , Issue.3 , pp. 332-384
  • 2
    • 84895729886 scopus 로고    scopus 로고
    • Clinical trials and late-stage drug development for Alzheimers disease: An appraisal from 1984 to 2014
    • Schneider LS, Mangialasche F, Andreasen N, et al. Clinical trials and late-stage drug development for Alzheimers disease: an appraisal from 1984 to 2014. J Intern Med. 2014; 275 (3): 251-283
    • (2014) J Intern Med , vol.275 , Issue.3 , pp. 251-283
    • Schneider, L.S.1    Mangialasche, F.2    Andreasen, N.3
  • 3
    • 77949768182 scopus 로고    scopus 로고
    • Towards disease-modifying treatment of Alzheimers disease: Drugs targeting beta-Amyloid
    • Frisardi V, Solfrizzi V, Imbimbo BP, et al. Towards disease-modifying treatment of Alzheimers disease: drugs targeting beta-Amyloid. Curr Alzheimer Res. 2010; 7 (1): 40-55
    • (2010) Curr Alzheimer Res , vol.7 , Issue.1 , pp. 40-55
    • Frisardi, V.1    Solfrizzi, V.2    Imbimbo, B.P.3
  • 4
    • 84907054900 scopus 로고    scopus 로고
    • Amyloid-directed monoclonal antibodies for the treatment of Alzheimers disease: The point of no return?
    • Panza F, Solfrizzi V, Imbimbo BP, et al. Amyloid-directed monoclonal antibodies for the treatment of Alzheimers disease: the point of no return? Expert Opin Biol Ther. 2014; 14 (10): 1465-1476
    • (2014) Expert Opin Biol Ther , vol.14 , Issue.10 , pp. 1465-1476
    • Panza, F.1    Solfrizzi, V.2    Imbimbo, B.P.3
  • 5
    • 84898059466 scopus 로고    scopus 로고
    • Tau-Aggregation inhibitor therapy for Alzheimers disease
    • Wischik CM, Harrington CR, Storey JM. Tau-Aggregation inhibitor therapy for Alzheimers disease. Biochem Pharmacol. 2014; 88 (4): 529-539
    • (2014) Biochem Pharmacol , vol.88 , Issue.4 , pp. 529-539
    • Wischik, C.M.1    Harrington, C.R.2    Storey, J.M.3
  • 6
    • 0030805991 scopus 로고    scopus 로고
    • Frequency of stages of Alzheimer-related lesions in different age categories
    • Braak H, Braak E. Frequency of stages of Alzheimer-related lesions in different age categories. Neurobiol Aging. 1997; 18 (4): 351-357
    • (1997) Neurobiol Aging , vol.18 , Issue.4 , pp. 351-357
    • Braak, H.1    Braak, E.2
  • 7
    • 84857558439 scopus 로고    scopus 로고
    • Immunotherapy for Alzheimers disease: From anti-β-Amyloid to tau-based immunization strategies
    • Panza F, Frisardi V, Solfrizzi V, et al. Immunotherapy for Alzheimers disease: from anti-β-Amyloid to tau-based immunization strategies. Immunotherapy. 2012; 4: 213-238
    • (2012) Immunotherapy , vol.4 , pp. 213-238
    • Panza, F.1    Frisardi, V.2    Solfrizzi, V.3
  • 8
    • 33947101532 scopus 로고    scopus 로고
    • A brief history of tau: The evolving view of the microtubule-Associated protein tau in neurodegenerative diseases
    • Lace GL, Wharton SB, Ince PG A brief history of tau: the evolving view of the microtubule-Associated protein tau in neurodegenerative diseases. Clin Neuropathol. 2007; 26 (2): 43-58
    • (2007) Clin Neuropathol , vol.26 , Issue.2 , pp. 43-58
    • Lace, G.L.1    Wharton, S.B.2    Ince, P.G.3
  • 9
    • 0003986552 scopus 로고
    • Isolation of a fragment of tau derived from the core of the paired helical filament of Alzheimers disease
    • Wischik CM, Novak M, Thøgersen HC, et al. Isolation of a fragment of tau derived from the core of the paired helical filament of Alzheimers disease. Proc Natl Acad Sci USA. 1988; 85 (12): 4506-4510
    • (1988) Proc Natl Acad Sci USA , vol.85 , Issue.12 , pp. 4506-4510
    • Wischik, C.M.1    Novak, M.2    Thøgersen, H.C.3
  • 10
    • 0025863618 scopus 로고
    • Neuropathological stageing of Alzheimer-related changes
    • Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 1991; 82 (4): 239-259
    • (1991) Acta Neuropathol , vol.82 , Issue.4 , pp. 239-259
    • Braak, H.1    Braak, E.2
  • 11
    • 0029686696 scopus 로고    scopus 로고
    • Evolution of the neuropathology of Alzheimers disease
    • Braak H, Braak E. Evolution of the neuropathology of Alzheimers disease. Acta Neurol Scand. 1996; 94 (S165): 3-12
    • (1996) Acta Neurol Scand , vol.94 , Issue.S165 , pp. 3-12
    • Braak, H.1    Braak, E.2
  • 12
    • 80054904670 scopus 로고    scopus 로고
    • Stages of the pathologic process in Alzheimer disease: Age categories from 1 to 100 years
    • Braak H, Thal DR, Ghebremedhin E, et al. Stages of the pathologic process in Alzheimer disease: age categories from 1 to 100 years. J Neuropathol Exp Neurol. 2011; 70 (11): 960-969
    • (2011) J Neuropathol Exp Neurol , vol.70 , Issue.11 , pp. 960-969
    • Braak, H.1    Thal, D.R.2    Ghebremedhin, E.3
  • 13
    • 0033899864 scopus 로고    scopus 로고
    • Staging of cytoskeletal and β-Amyloid changes in human isocortex reveals biphasic synaptic protein response during progression of Alzheimers disease
    • Mukaetova-Ladinska EB, Garcia-Siera F, Hurt J, et al. Staging of cytoskeletal and β-Amyloid changes in human isocortex reveals biphasic synaptic protein response during progression of Alzheimers disease. Am J Pathol. 2000; 157 (2): 623-636
    • (2000) Am J Pathol , vol.157 , Issue.2 , pp. 623-636
    • Mukaetova-Ladinska, E.B.1    Garcia-Siera, F.2    Hurt, J.3
  • 14
    • 79451470073 scopus 로고    scopus 로고
    • Tau pathology in children and young adults: Can you still be unconditionally baptist?
    • Duyckaerts C. Tau pathology in children and young adults: can you still be unconditionally baptist? Acta Neuropathol. 2011; 121 (2): 145-147
    • (2011) Acta Neuropathol , vol.121 , Issue.2 , pp. 145-147
    • Duyckaerts, C.1
  • 15
    • 0033394155 scopus 로고    scopus 로고
    • Memory and mental status correlates of modified Braak staging
    • Grober E, Dickson D, Sliwinski MJ, et al. Memory and mental status correlates of modified Braak staging. Neurobiol Aging. 1999; 20 (6): 573-579
    • (1999) Neurobiol Aging , vol.20 , Issue.6 , pp. 573-579
    • Grober, E.1    Dickson, D.2    Sliwinski, M.J.3
  • 16
    • 84860215480 scopus 로고    scopus 로고
    • Correlation of Alzheimer disease neuropathologic changes with cognitive status: A review of the literature
    • Nelson PT, Alafuzoff I, Bigio EH, et al. Correlation of Alzheimer disease neuropathologic changes with cognitive status: a review of the literature. J Neuropathol Exp Neurol. 2012; 71 (5): 362-381
    • (2012) J Neuropathol Exp Neurol , vol.71 , Issue.5 , pp. 362-381
    • Nelson, P.T.1    Alafuzoff, I.2    Bigio, E.H.3
  • 17
    • 0346124139 scopus 로고    scopus 로고
    • Cerebrospinal fluid tau and beta-Amyloid: How well do these biomarkers reflect autopsy-confirmed dementia diagnoses?
    • Clark CM, Xie S, Chittams J, et al. Cerebrospinal fluid tau and beta-Amyloid: how well do these biomarkers reflect autopsy-confirmed dementia diagnoses? Arch Neurol. 2003; 60 (12): 1696-1702
    • (2003) Arch Neurol , vol.60 , Issue.12 , pp. 1696-1702
    • Clark, C.M.1    Xie, S.2    Chittams, J.3
  • 18
    • 84872379077 scopus 로고    scopus 로고
    • Tracking pathophysiological processes in Alzheimers disease: An updated hypothetical model of dynamic biomarkers
    • Jack CR Jr, Knopman DS, Jagust WJ, et al. Tracking pathophysiological processes in Alzheimers disease: an updated hypothetical model of dynamic biomarkers. Lancet Neurol. 2013; 12 (2): 207-216
    • (2013) Lancet Neurol , vol.12 , Issue.2 , pp. 207-216
    • Jack, C.R.1    Knopman, D.S.2    Jagust, W.J.3
  • 19
    • 84865529158 scopus 로고    scopus 로고
    • Clinical and biomarker changes in dominantly inherited Alzheimers disease
    • Bateman RJ, Xiong C, Benzinger TL, et al. Clinical and biomarker changes in dominantly inherited Alzheimers disease. N Engl J Med. 2012; 367: 795-804
    • (2012) N Engl J Med , vol.367 , pp. 795-804
    • Bateman, R.J.1    Xiong, C.2    Benzinger, T.L.3
  • 20
    • 84906691088 scopus 로고    scopus 로고
    • A data-driven model of biomarker changes in sporadic Alzheimers disease
    • Young AL, Oxtoby NP, Daga P, et al A data-driven model of biomarker changes in sporadic Alzheimers disease. Brain. 2014; 137 (Pt 9): 2564-2577
    • (2014) Brain , vol.137 , pp. 2564-2577
    • Young, A.L.1    Oxtoby, N.P.2    Daga, P.3
  • 21
    • 72049130805 scopus 로고    scopus 로고
    • Hypothetical model of dynamic biomarkers of the Alzheimers pathological cascade
    • Jack CR, Knopman DS, Jagust WJ, et al. Hypothetical model of dynamic biomarkers of the Alzheimers pathological cascade. Lancet Neurol. 2010; 9: 119-128
    • (2010) Lancet Neurol , vol.9 , pp. 119-128
    • Jack, C.R.1    Knopman, D.S.2    Jagust, W.J.3
  • 22
    • 77955480521 scopus 로고    scopus 로고
    • Clinical core of the Alzheimers disease neuroimaging initiative: Progress and plans
    • Aisen PS, Petersen RC, Donohue MC, et al. Clinical core of the Alzheimers disease neuroimaging initiative: progress and plans. Alzheimers Dement. 2010; 6 (3): 239-246
    • (2010) Alzheimers Dement , vol.6 , Issue.3 , pp. 239-246
    • Aisen, P.S.1    Petersen, R.C.2    Donohue, M.C.3
  • 23
    • 0022744803 scopus 로고
    • Abnormal phosphorylation of the microtubule-Associated protein tau (tau) in Alzheimer cytoskeletal pathology
    • Grundke-Iqbal I, Iqbal K, Tung YC, et al. Abnormal phosphorylation of the microtubule-Associated protein tau (tau) in Alzheimer cytoskeletal pathology. Proc Natl Acad Sci U S A. 1986; 83 (13): 4913-4917
    • (1986) Proc Natl Acad Sci U S A , vol.83 , Issue.13 , pp. 4913-4917
    • Grundke-Iqbal, I.1    Iqbal, K.2    Tung, Y.C.3
  • 24
    • 0026595846 scopus 로고
    • Tau proteins of Alzheimer paired helical filaments: Abnormal phosphorylation of all six brain isoforms
    • Goedert M, Spillantini MG, Cairns NJ, et al. Tau proteins of Alzheimer paired helical filaments: abnormal phosphorylation of all six brain isoforms. Neuron. 1992; 8 (1): 159-168
    • (1992) Neuron , vol.8 , Issue.1 , pp. 159-168
    • Goedert, M.1    Spillantini, M.G.2    Cairns, N.J.3
  • 25
    • 0024044195 scopus 로고
    • Structural characterization of the core of the paired helical filament of Alzheimer disease
    • Jul
    • Wischik CM, Novak M, Edwards PC, et al. Structural characterization of the core of the paired helical filament of Alzheimer disease. Proc Natl Acad Sci U S A. 1988 Jul; 85 (13): 4884-4888
    • (1988) Proc Natl Acad Sci U S A , vol.85 , Issue.13 , pp. 4884-4888
    • Wischik, C.M.1    Novak, M.2    Edwards, P.C.3
  • 26
    • 0027398169 scopus 로고
    • Molecular characterization of the minimal protease resistant tau unit of the Alzheimers disease paired helical filament
    • Novak M, Kabat J, Wischik CM. Molecular characterization of the minimal protease resistant tau unit of the Alzheimers disease paired helical filament. EMBO J. 1993; 12 (1): 365-370
    • (1993) EMBO J , vol.12 , Issue.1 , pp. 365-370
    • Novak, M.1    Kabat, J.2    Wischik, C.M.3
  • 27
    • 20044381844 scopus 로고    scopus 로고
    • Tau truncation during neurofibrillary tangle evolution in Alzheimers disease
    • Guillozet-Bongaarts AL, Garcia-Sierra F, Reynolds MR, et al. Tau truncation during neurofibrillary tangle evolution in Alzheimers disease. Neurobiol Aging. 2005; 26 (7): 1015-1022
    • (2005) Neurobiol Aging , vol.26 , Issue.7 , pp. 1015-1022
    • Guillozet-Bongaarts, A.L.1    Garcia-Sierra, F.2    Reynolds, M.R.3
  • 28
    • 0028023944 scopus 로고
    • Glycated tau protein in Alzheimer disease: A mechanism for induction of oxidant stress
    • Yan SD, Chen X, Schmidt AM, et al. Glycated tau protein in Alzheimer disease: a mechanism for induction of oxidant stress. Proc Natl Acad Sci U S A. 1994; 91 (16): 7787-7791
    • (1994) Proc Natl Acad Sci U S A , vol.91 , Issue.16 , pp. 7787-7791
    • Yan, S.D.1    Chen, X.2    Schmidt, A.M.3
  • 29
    • 0029133373 scopus 로고
    • Tau protein from Alzheimers disease patients is glycated at its tubulin-binding domain
    • Ledesma MD, Bonay P, Avila J. Tau protein from Alzheimers disease patients is glycated at its tubulin-binding domain. J Neurochem. 1995; 65 (4): 1658-1664
    • (1995) J Neurochem , vol.65 , Issue.4 , pp. 1658-1664
    • Ledesma, M.D.1    Bonay, P.2    Avila, J.3
  • 30
    • 13444287877 scopus 로고    scopus 로고
    • Site-specific nitration and oxidative dityrosine bridging of the tau protein by peroxynitrite: Implications for Alzheimers disease
    • Reynolds MR, Berry RW, Binder LI. Site-specific nitration and oxidative dityrosine bridging of the tau protein by peroxynitrite: implications for Alzheimers disease. Biochemistry. 2005; 44 (5): 1690-1700
    • (2005) Biochemistry , vol.44 , Issue.5 , pp. 1690-1700
    • Reynolds, M.R.1    Berry, R.W.2    Binder, L.I.3
  • 31
    • 33750940057 scopus 로고    scopus 로고
    • Tau nitration occurs at tyrosine 29 in the fibrillar lesions of Alzheimers disease and other tauopathies
    • Reynolds MR, Reyes JF, Fu Y, et al. Tau nitration occurs at tyrosine 29 in the fibrillar lesions of Alzheimers disease and other tauopathies. J Neurosci. 2006; 26 (42): 10636-10645
    • (2006) J Neurosci , vol.26 , Issue.42 , pp. 10636-10645
    • Reynolds, M.R.1    Reyes, J.F.2    Fu, Y.3
  • 32
    • 68949212379 scopus 로고    scopus 로고
    • Lysine acetylation targets protein complexes and co-regulates major cellular functions
    • Choudhary C, Kumar C, Gnad F, et al. Lysine acetylation targets protein complexes and co-regulates major cellular functions. Science. 2009; 325 (5942): 834-840
    • (2009) Science , vol.325 , Issue.5942 , pp. 834-840
    • Choudhary, C.1    Kumar, C.2    Gnad, F.3
  • 33
    • 77957001697 scopus 로고    scopus 로고
    • Acetylation of tau inhibits its degradation and contributes to tauopathy
    • Min S-W, Cho S-H, Zhou Y, et al. Acetylation of tau inhibits its degradation and contributes to tauopathy. Neuron. 2010; 67 (6): 953-966
    • (2010) Neuron , vol.67 , Issue.6 , pp. 953-966
    • Min, S.-W.1    Cho, S.-H.2    Zhou, Y.3
  • 34
    • 10544236116 scopus 로고    scopus 로고
    • The microtubule-Associated protein tau is extensively modified with O-linked N-Acetylglucosamine
    • Arnold CS, Johnson GV, Cole RN, et al. The microtubule-Associated protein tau is extensively modified with O-linked N-Acetylglucosamine. J Biol Chem. 1996; 271 (46): 28741-28744
    • (1996) J Biol Chem , vol.271 , Issue.46 , pp. 28741-28744
    • Arnold, C.S.1    Johnson, G.V.2    Cole, R.N.3
  • 35
    • 3242739968 scopus 로고    scopus 로고
    • O-GlcNAcylation regulates phosphorylation of tau: A mechanism involved in Alzheimers disease
    • Liu F, Iqbal K, Grundke-Iqbal I, et al. O-GlcNAcylation regulates phosphorylation of tau: a mechanism involved in Alzheimers disease. Proc Natl Acad Sci USA. 2004; 101 (29): 10804-10809
    • (2004) Proc Natl Acad Sci USA , vol.101 , Issue.29 , pp. 10804-10809
    • Liu, F.1    Iqbal, K.2    Grundke-Iqbal, I.3
  • 36
    • 0024560685 scopus 로고
    • Immunochemical properties of ubiquitin conjugates in the paired helical filaments of Alzheimer disease
    • Perry G, Mulvihill P, Fried VA, et al. Immunochemical properties of ubiquitin conjugates in the paired helical filaments of Alzheimer disease. J Neurochem. 1989; 52 (5): 1523-1528
    • (1989) J Neurochem , vol.52 , Issue.5 , pp. 1523-1528
    • Perry, G.1    Mulvihill, P.2    Fried, V.A.3
  • 37
    • 0026298636 scopus 로고
    • Ubiquitination and abnormal phosphorylation of paired helical filaments in Alzheimers disease
    • Iqbal K, Grundke-Iqbal I. Ubiquitination and abnormal phosphorylation of paired helical filaments in Alzheimers disease. Mol Neurobiol. 1991; 5 (2-4): 399-410
    • (1991) Mol Neurobiol , vol.5 , Issue.24 , pp. 399-410
    • Iqbal, K.1    Grundke-Iqbal, I.2
  • 38
    • 84862300056 scopus 로고    scopus 로고
    • Structure and pathology of tau protein in Alzheimer disease
    • Kolarova M, García-Sierra F, Bartos A, et al. Structure and pathology of tau protein in Alzheimer disease. Int J Alzheimers Dis. 2012; 2012: 731526
    • (2012) Int J Alzheimers Dis , vol.2012 , pp. 731526
    • Kolarova, M.1    García-Sierra, F.2    Bartos, A.3
  • 39
    • 67349191008 scopus 로고    scopus 로고
    • Truncated tau at D421 is associated with neurodegeneration and tangle formation in the brain of Alzheimer transgenic models
    • Zhang Q, Zhang X, Sun A. Truncated tau at D421 is associated with neurodegeneration and tangle formation in the brain of Alzheimer transgenic models. Acta Neuropathol. 2009; 117 (6): 687-697
    • (2009) Acta Neuropathol , vol.117 , Issue.6 , pp. 687-697
    • Zhang, Q.1    Zhang, X.2    Sun, A.3
  • 40
    • 77953544031 scopus 로고    scopus 로고
    • Tau phosphorylation and cleavage in ethanol-induced neurodegeneration in the developing mouse brain
    • Saito M, Chakraborty G, Mao R-F, et al. Tau phosphorylation and cleavage in ethanol-induced neurodegeneration in the developing mouse brain. Neurochem Res. 2010; 35 (4): 651-659
    • (2010) Neurochem Res , vol.35 , Issue.4 , pp. 651-659
    • Saito, M.1    Chakraborty, G.2    Mao, R.-F.3
  • 41
    • 79953087890 scopus 로고    scopus 로고
    • The acetylation of tau inhibits its function and promotes pathological tau aggregation
    • Cohen TJ, Guo JL, Hurtado DE, et al. The acetylation of tau inhibits its function and promotes pathological tau aggregation. Nat Commun. 2011; 2: 252
    • (2011) Nat Commun , vol.2 , pp. 252
    • Cohen, T.J.1    Guo, J.L.2    Hurtado, D.E.3
  • 42
    • 0032747486 scopus 로고    scopus 로고
    • Activation of caspase-3 in single neurons and autophagic granules of granulovacuolar degeneration in Alzheimers disease Evidence for apoptotic cell death
    • Stadelmann C, Deckwerth TL, Srinivasan A, et al. Activation of caspase-3 in single neurons and autophagic granules of granulovacuolar degeneration in Alzheimers disease. Evidence for apoptotic cell death. Am J Pathol. 1999; 155 (2): 1459-1466
    • (1999) Am J Pathol , vol.155 , Issue.2 , pp. 1459-1466
    • Stadelmann, C.1    Deckwerth, T.L.2    Srinivasan, A.3
  • 43
    • 0036968953 scopus 로고    scopus 로고
    • Caspase-9 activation and caspase cleavage of tau in the Alzheimers disease brain
    • Rohn TT, Rissman RA, Davis MC, et al. Caspase-9 activation and caspase cleavage of tau in the Alzheimers disease brain. Neurobiol Dis. 2002; 11 (2): 341-354
    • (2002) Neurobiol Dis , vol.11 , Issue.2 , pp. 341-354
    • Rohn, T.T.1    Rissman, R.A.2    Davis, M.C.3
  • 44
    • 78649660787 scopus 로고    scopus 로고
    • Aggregation of detergent-insoluble tau is involved in neuronal loss but not in synaptic loss
    • Kimura T, Fukuda T, Sahara N, et al. Aggregation of detergent-insoluble tau is involved in neuronal loss but not in synaptic loss. J Biol Chem. 2010; 285 (49): 38692-38699
    • (2010) J Biol Chem , vol.285 , Issue.49 , pp. 38692-38699
    • Kimura, T.1    Fukuda, T.2    Sahara, N.3
  • 45
    • 79960325654 scopus 로고    scopus 로고
    • The toxicity of tau in Alzheimer disease: Turnover, targets and potential therapeutics
    • Pritchard SM, Dolan PJ, Vitkus A, et al. The toxicity of tau in Alzheimer disease: turnover, targets and potential therapeutics. J Cell Mol Med. 2011; 15 (8): 1621-1635
    • (2011) J Cell Mol Med , vol.15 , Issue.8 , pp. 1621-1635
    • Pritchard, S.M.1    Dolan, P.J.2    Vitkus, A.3
  • 46
    • 7244236320 scopus 로고    scopus 로고
    • Inclusion body formation reduces levels of mutant huntingtin and the risk of neuronal death
    • Arrasate M, Mitra S, Schweitzer ES, et al. Inclusion body formation reduces levels of mutant huntingtin and the risk of neuronal death. Nature. 2004; 431 (7010): 805-810
    • (2004) Nature , vol.431 , Issue.7010 , pp. 805-810
    • Arrasate, M.1    Mitra, S.2    Schweitzer, E.S.3
  • 47
    • 33750363298 scopus 로고    scopus 로고
    • The roles of intracellular protein-degradation pathways in neurodegeneration
    • Rubinsztein DC. The roles of intracellular protein-degradation pathways in neurodegeneration. Nature. 2006; 443 (7113): 780-786
    • (2006) Nature , vol.443 , Issue.7113 , pp. 780-786
    • Rubinsztein, D.C.1
  • 48
    • 67649273927 scopus 로고    scopus 로고
    • Propagation of tau misfolding from the outside to the inside of a cell
    • Frost B, Jacks RL, Diamond MI. Propagation of tau misfolding from the outside to the inside of a cell. J Biol Chem. 2009; 284 (19): 12845-12852
    • (2009) J Biol Chem , vol.284 , Issue.19 , pp. 12845-12852
    • Frost, B.1    Jacks, R.L.2    Diamond, M.I.3
  • 49
    • 67650077008 scopus 로고    scopus 로고
    • Transmission and spreading of tauopathy in transgenic mouse brain
    • Clavaguera F, Bolmont T, Crowther RA, et al. Transmission and spreading of tauopathy in transgenic mouse brain. Nat Cell Biol. 2009; 11 (7): 909-913
    • (2009) Nat Cell Biol , vol.11 , Issue.7 , pp. 909-913
    • Clavaguera, F.1    Bolmont, T.2    Crowther, R.A.3
  • 50
    • 84857275902 scopus 로고    scopus 로고
    • Propagation of tau pathology in a model of early Alzheimers disease
    • de Calignon A, Polydoro M, Suárez-Calvet M, et al. Propagation of tau pathology in a model of early Alzheimers disease. Neuron. 2012; 73 (4): 685-697
    • (2012) Neuron , vol.73 , Issue.4 , pp. 685-697
    • De Calignon, A.1    Polydoro, M.2    Suárez-Calvet, M.3
  • 51
    • 34548146119 scopus 로고    scopus 로고
    • Immunotherapy targeting pathological tau conformers in a tangle mouse model reduces brain pathology with associated functional improvements
    • Asuni AA, Boutajangout A, Quartermain D, et al. Immunotherapy targeting pathological tau conformers in a tangle mouse model reduces brain pathology with associated functional improvements. J Neurosci. 2007; 27 (34): 9115-9129
    • (2007) J Neurosci , vol.27 , Issue.34 , pp. 9115-9129
    • Asuni, A.A.1    Boutajangout, A.2    Quartermain, D.3
  • 52
    • 20444413356 scopus 로고    scopus 로고
    • Effects of α-synuclein immunization in a mouse model of Parkinsons disease
    • Masliah E, Rockenstein E, Adame A, et al. Effects of α-synuclein immunization in a mouse model of Parkinsons disease. Neuron. 2005; 46 (6): 857-868
    • (2005) Neuron , vol.46 , Issue.6 , pp. 857-868
    • Masliah, E.1    Rockenstein, E.2    Adame, A.3
  • 53
    • 33847662852 scopus 로고    scopus 로고
    • Soluble protein oligomers in neurodegeneration: Lessons from the Alzheimers amyloid β-peptide
    • Haass C, Selkoe DJ. Soluble protein oligomers in neurodegeneration: lessons from the Alzheimers amyloid β-peptide. Nat Rev Mol Cell Biol. 2007; 8 (2): 101-112
    • (2007) Nat Rev Mol Cell Biol , vol.8 , Issue.2 , pp. 101-112
    • Haass, C.1    Selkoe, D.J.2
  • 54
    • 67349142329 scopus 로고    scopus 로고
    • Mechanisms of tau-induced neurodegeneration
    • Iqbal K, Liu F, Gong C-X, et al. Mechanisms of tau-induced neurodegeneration. Acta Neuropathol. 2009; 118 (1): 53-69
    • (2009) Acta Neuropathol , vol.118 , Issue.1 , pp. 53-69
    • Iqbal, K.1    Liu, F.2    Gong, C.-X.3
  • 55
    • 70349638299 scopus 로고    scopus 로고
    • Advances in tau-focused drug discovery for Alzheimers disease and related tauopathies
    • Brunden KR, Trojanowski JQ, Lee VM. Advances in tau-focused drug discovery for Alzheimers disease and related tauopathies. Nat Rev Drug Discov. 2009; 8 (10): 783-793
    • (2009) Nat Rev Drug Discov , vol.8 , Issue.10 , pp. 783-793
    • Brunden, K.R.1    Trojanowski, J.Q.2    Lee, V.M.3
  • 56
    • 20044367108 scopus 로고    scopus 로고
    • Cell-cycle reentry and cell death in transgenic mice expressing nonmutant human tau isoforms
    • Andorfer C, Acker CM, Kress Y, et al. Cell-cycle reentry and cell death in transgenic mice expressing nonmutant human tau isoforms. J Neurosci. 2005; 25 (22): 5446-5454
    • (2005) J Neurosci , vol.25 , Issue.22 , pp. 5446-5454
    • Andorfer, C.1    Acker, C.M.2    Kress, Y.3
  • 57
    • 68249138795 scopus 로고    scopus 로고
    • Tangle-bearing neurons survive despite disruption of membrane integrity in a mouse model of tauopathy
    • de Calignon A, Spires-Jones TL, Pitstick R, et al. Tangle-bearing neurons survive despite disruption of membrane integrity in a mouse model of tauopathy. J Neuropathol Exp Neurol. 2009; 68 (7): 757-761
    • (2009) J Neuropathol Exp Neurol , vol.68 , Issue.7 , pp. 757-761
    • De Calignon, A.1    Spires-Jones, T.L.2    Pitstick, R.3
  • 58
    • 34248181511 scopus 로고    scopus 로고
    • Reducing endogenous tau ameliorates amyloid beta-induced deficits in an Alzheimers disease mouse model
    • Roberson ED, Scearce-Levie K, Palop JJ, et al. Reducing endogenous tau ameliorates amyloid beta-induced deficits in an Alzheimers disease mouse model. Science. 2007; 316 (5825): 750-754
    • (2007) Science , vol.316 , Issue.5825 , pp. 750-754
    • Roberson, E.D.1    Scearce-Levie, K.2    Palop, J.J.3
  • 59
    • 77957737750 scopus 로고    scopus 로고
    • Tau reduction prevents A{beta}-induced defects in axonal transport
    • Vossel KA, Zhang K, Brodbeck J, et al. Tau reduction prevents A{beta}-induced defects in axonal transport. Science. 2010; 330 (6001): 198
    • (2010) Science , vol.330 , Issue.6001 , pp. 198
    • Vossel, K.A.1    Zhang, K.2    Brodbeck, J.3
  • 60
    • 73949142307 scopus 로고    scopus 로고
    • Amyloid-beta and tau synergistically impair the oxidative phosphorylation system in triple transgenic Alzheimers disease mice
    • Rhein V, Song X, Wiesner A, et al. Amyloid-beta and tau synergistically impair the oxidative phosphorylation system in triple transgenic Alzheimers disease mice. Proc Natl Acad Sci USA. 2009; 106 (47): 20057-20062
    • (2009) Proc Natl Acad Sci USA , vol.106 , Issue.47 , pp. 20057-20062
    • Rhein, V.1    Song, X.2    Wiesner, A.3
  • 61
    • 33846118789 scopus 로고    scopus 로고
    • Multiple-motor based transport and its regulation by tau
    • Vershinin M, Carter BC, Razafsky DS, et al. Multiple-motor based transport and its regulation by tau. Proc Natl Acad Sci USA. 2007; 104 (1): 87-92
    • (2007) Proc Natl Acad Sci USA , vol.104 , Issue.1 , pp. 87-92
    • Vershinin, M.1    Carter, B.C.2    Razafsky, D.S.3
  • 62
    • 77952548853 scopus 로고    scopus 로고
    • Glycogen synthase kinase-3β and the p25 activator of cyclin dependent kinase 5 increase pausing of mitochondria in neurons
    • Morel M, Authelet M, Dedecker R, et al. Glycogen synthase kinase-3β and the p25 activator of cyclin dependent kinase 5 increase pausing of mitochondria in neurons. Neuroscience. 2010; 167 (4): 1044-1056
    • (2010) Neuroscience , vol.167 , Issue.4 , pp. 1044-1056
    • Morel, M.1    Authelet, M.2    Dedecker, R.3
  • 63
    • 57349172663 scopus 로고    scopus 로고
    • Parkinsonism and impaired axonal transport in a mouse model of frontotemporal dementia
    • Ittner LM, Fath T, Ke YD, et al. Parkinsonism and impaired axonal transport in a mouse model of frontotemporal dementia. Proc Natl Acad Sci USA. 2008; 105 (41): 15997-16002
    • (2008) Proc Natl Acad Sci USA , vol.105 , Issue.41 , pp. 15997-16002
    • Ittner, L.M.1    Fath, T.2    Ke, Y.D.3
  • 64
    • 68949105821 scopus 로고    scopus 로고
    • Phosphorylated tau interacts with c-Jun N-Terminal kinase interacting protein 1 (JIP1) in Alzheimer disease
    • Ittner LM, Ke YD, Gotz J. Phosphorylated tau interacts with c-Jun N-Terminal kinase interacting protein 1 (JIP1) in Alzheimer disease. J Biol Chem. 2009; 284 (31): 20909-20916
    • (2009) J Biol Chem , vol.284 , Issue.31 , pp. 20909-20916
    • Ittner, L.M.1    Ke, Y.D.2    Gotz, J.3
  • 65
    • 77955089472 scopus 로고    scopus 로고
    • Convergence of amyloid-β and Tau pathologies on mitochondria in vivo
    • Eckert A, Schulz KL, Rhein V, et al. Convergence of amyloid-β and Tau pathologies on mitochondria in vivo. Mol Neurobiol. 2010; 41 (2-3): 107-114
    • (2010) Mol Neurobiol , vol.41 , Issue.23 , pp. 107-114
    • Eckert, A.1    Schulz, K.L.2    Rhein, V.3
  • 66
    • 84867608068 scopus 로고    scopus 로고
    • Tau protein kinases: Involvement in Alzheimers disease
    • Martin L, Latypova X, Wilson CM, et al. Tau protein kinases: involvement in Alzheimers disease. Ageing Res Rev. 2013; 12 (1): 289-309
    • (2013) Ageing Res Rev , vol.12 , Issue.1 , pp. 289-309
    • Martin, L.1    Latypova, X.2    Wilson, C.M.3
  • 67
    • 84894223460 scopus 로고    scopus 로고
    • Microtubule-Associated protein tau as a therapeutic target in Alzheimers disease
    • Iqbal K, Gong C-X, Liu F. Microtubule-Associated protein tau as a therapeutic target in Alzheimers disease. Expert Opin Ther Targets. 2014; 18 (3): 307-318
    • (2014) Expert Opin Ther Targets , vol.18 , Issue.3 , pp. 307-318
    • Iqbal, K.1    Gong, C.-X.2    Liu, F.3
  • 68
    • 84930538607 scopus 로고    scopus 로고
    • Tau immunotherapy for alzheimers disease
    • Pedersen JT, Sigurdsson EM. Tau immunotherapy for Alzheimers disease. Trends Mol Med. 2015; 21 (6): 394-402
    • (2015) Trends Mol Med , vol.21 , Issue.6 , pp. 394-402
    • Pedersen, J.T.1    Sigurdsson, E.M.2
  • 69
    • 84941600642 scopus 로고    scopus 로고
    • Early investigational drugs targeting tau protein for the treatment of Alzheimers disease
    • Anand K, Sabbagh M. Early investigational drugs targeting tau protein for the treatment of Alzheimers disease. Expert Opin Investig Drugs. 2015; 24 (10): 1355-1360
    • (2015) Expert Opin Investig Drugs , vol.24 , Issue.10 , pp. 1355-1360
    • Anand, K.1    Sabbagh, M.2
  • 70
    • 0026694783 scopus 로고
    • Brain levels of microtubule-Associated protein tau are elevated in Alzheimers disease: A radioimmuno-slot-blot assay for nanograms of the protein
    • Khatoon S, Grundke-Iqbal I, Iqbal K. Brain levels of microtubule-Associated protein tau are elevated in Alzheimers disease: a radioimmuno-slot-blot assay for nanograms of the protein. J Neurochem. 1992; 59 (2): 750-753
    • (1992) J Neurochem , vol.59 , Issue.2 , pp. 750-753
    • Khatoon, S.1    Grundke-Iqbal, I.2    Iqbal, K.3
  • 71
    • 40449087334 scopus 로고    scopus 로고
    • Interplay between cyclin-dependent kinase 5 and glycogen synthase kinase 3b mediated by neuregulin signaling leads to differential effects on tau phosphorylation and amyloid precursor protein processing
    • Wen Y, Planel E, Herman M, et al. Interplay between cyclin-dependent kinase 5 and glycogen synthase kinase 3b mediated by neuregulin signaling leads to differential effects on tau phosphorylation and amyloid precursor protein processing. J Neurosci. 2008; 28 (10): 2624-2632
    • (2008) J Neurosci , vol.28 , Issue.10 , pp. 2624-2632
    • Wen, Y.1    Planel, E.2    Herman, M.3
  • 72
    • 84898056918 scopus 로고    scopus 로고
    • A phase 2 trial of the GSK-3 inhibitor tideglusib in progressive supranuclear palsy
    • Tolosa E, Litvan I, Höglinger GU, et al A phase 2 trial of the GSK-3 inhibitor tideglusib in progressive supranuclear palsy. Mov Disord. 2014; 29 (4): 470-478
    • (2014) Mov Disord , vol.29 , Issue.4 , pp. 470-478
    • Tolosa, E.1    Litvan, I.2    Höglinger, G.U.3
  • 73
    • 84872470005 scopus 로고    scopus 로고
    • Treatment of Alzheimers disease with the GSK-3 inhibitor tideglusib: A pilot study
    • Del Ser T, Steinwachs KC, Gertz HJ, et al. Treatment of Alzheimers disease with the GSK-3 inhibitor tideglusib: a pilot study. J Alzheimers Dis. 2013; 33 (1): 205-215
    • (2013) J Alzheimers Dis , vol.33 , Issue.1 , pp. 205-215
    • Del Ser, T.1    Steinwachs, K.C.2    Gertz, H.J.3
  • 74
    • 84924106368 scopus 로고    scopus 로고
    • A phase II trial of tideglusib in Alzheimers disease
    • Lovestone S, Boada M, Dubois B, et al A phase II trial of tideglusib in Alzheimers disease. J Alzheimers Dis. 2015; 45 (1): 75-88
    • (2015) J Alzheimers Dis , vol.45 , Issue.1 , pp. 75-88
    • Lovestone, S.1    Boada, M.2    Dubois, B.3
  • 75
    • 0035851175 scopus 로고    scopus 로고
    • Reduced protein phosphatase 2A activity induces hyperphosphorylation and altered compartmentalization of tau in transgenic mice
    • Kins S, Crameri A, Evans DR, et al. Reduced protein phosphatase 2A activity induces hyperphosphorylation and altered compartmentalization of tau in transgenic mice. J Biol Chem. 2001; 276 (41): 38193-38200
    • (2001) J Biol Chem , vol.276 , Issue.41 , pp. 38193-38200
    • Kins, S.1    Crameri, A.2    Evans, D.R.3
  • 76
    • 79958744416 scopus 로고    scopus 로고
    • Phosphoprotein phosphatase 2A: A novel druggable target for Alzheimers disease
    • Voronkov M, Braithwaite SP, Stock JB. Phosphoprotein phosphatase 2A: a novel druggable target for Alzheimers disease. Future Med Chem. 2011; 3 (7): 821-833
    • (2011) Future Med Chem , vol.3 , Issue.7 , pp. 821-833
    • Voronkov, M.1    Braithwaite, S.P.2    Stock, J.B.3
  • 77
    • 84920765954 scopus 로고    scopus 로고
    • Further understanding of tau phosphorylation: Implications for therapy
    • Medina M, Avila J. Further understanding of tau phosphorylation: implications for therapy. Expert Rev Neurother. 2015; 15 (1): 115-122
    • (2015) Expert Rev Neurother , vol.15 , Issue.1 , pp. 115-122
    • Medina, M.1    Avila, J.2
  • 78
    • 78649859760 scopus 로고    scopus 로고
    • The carboxy-Terminal fragment of inhibitor-2 of protein phosphatase-2A induces Alzheimer disease pathology and cognitive impairment
    • Wang X, Blanchard J, Kohlbrenner E, et al. The carboxy-Terminal fragment of inhibitor-2 of protein phosphatase-2A induces Alzheimer disease pathology and cognitive impairment. FASEB J. 2010; 24 (11): 4420-321
    • (2010) FASEB J , vol.24 , Issue.11 , pp. 4420-5321
    • Wang, X.1    Blanchard, J.2    Kohlbrenner, E.3
  • 79
    • 79959534777 scopus 로고    scopus 로고
    • Molecular implication of PP2A and Pin1 in the Alzheimers disease specific hyperphosphorylation of Tau
    • Landrieu I, Smet-Nocca C, Amniai L, et al. Molecular implication of PP2A and Pin1 in the Alzheimers disease specific hyperphosphorylation of Tau. PLoS One. 2011; 6 (6): e21521
    • (2011) PLoS One , vol.6 , Issue.6 , pp. e21521
    • Landrieu, I.1    Smet-Nocca, C.2    Amniai, L.3
  • 80
    • 84884537540 scopus 로고    scopus 로고
    • Leucine carboxyl methyltransferase 1 (LCMT1)-dependent methylation regulates the association of protein phosphatase 2A and Tau protein with plasma membrane microdomains in neuroblastoma cells
    • Sontag J-M, Nunbhakdi-Craig V, Sontag E. Leucine carboxyl methyltransferase 1 (LCMT1)-dependent methylation regulates the association of protein phosphatase 2A and Tau protein with plasma membrane microdomains in neuroblastoma cells. J Biol Chem. 2013; 288 (38): 27396-27405
    • (2013) J Biol Chem , vol.288 , Issue.38 , pp. 27396-27405
    • Sontag, J.-M.1    Nunbhakdi-Craig, V.2    Sontag, E.3
  • 81
    • 33947519211 scopus 로고    scopus 로고
    • Microtubule-stabilizing agent prevents protein accumulation-induced loss of synaptic markers
    • Butler D, Bendiske J, Michaelis M, et al. Microtubule-stabilizing agent prevents protein accumulation-induced loss of synaptic markers. Eur J Pharmacol. 2007; 562 (1-2): 20-27
    • (2007) Eur J Pharmacol , vol.562 , Issue.12 , pp. 20-27
    • Butler, D.1    Bendiske, J.2    Michaelis, M.3
  • 82
    • 84863230105 scopus 로고    scopus 로고
    • The microtubule-stabilizing agent, epothilone D, reduces axonal dysfunction, neurotoxicity, cognitive deficits, and Alzheimer-like pathology in an interventional study with aged tau transgenic mice
    • Zhang B, Carroll J, Trojanowski JQ, et al. The microtubule-stabilizing agent, epothilone D, reduces axonal dysfunction, neurotoxicity, cognitive deficits, and Alzheimer-like pathology in an interventional study with aged tau transgenic mice. J Neurosci. 2012; 32 (11): 3601-3611
    • (2012) J Neurosci , vol.32 , Issue.11 , pp. 3601-3611
    • Zhang, B.1    Carroll, J.2    Trojanowski, J.Q.3
  • 84
    • 0033049956 scopus 로고    scopus 로고
    • Complete sequence of a novel protein containing a femtomolar-Activity-dependent neuroprotective peptide
    • Bassan M, Zamostiano R, et al. Complete sequence of a novel protein containing a femtomolar-Activity-dependent neuroprotective peptide. J Neurochem. 1999; 72 (3): 1283-1293
    • (1999) J Neurochem , vol.72 , Issue.3 , pp. 1283-1293
    • Bassan, M.1    Zamostiano, R.2
  • 85
    • 35548974346 scopus 로고    scopus 로고
    • Activity-dependent neuroprotective protein snippet NAP reduces tau hyperphosphorylation and enhances learning in a novel transgenic mouse model
    • Vulih-Shultzman I, Pinhasov A, Mandel S, et al. Activity-dependent neuroprotective protein snippet NAP reduces tau hyperphosphorylation and enhances learning in a novel transgenic mouse model. J Pharmacol Exp Ther. 2007; 323 (2): 438-449
    • (2007) J Pharmacol Exp Ther , vol.323 , Issue.2 , pp. 438-449
    • Vulih-Shultzman, I.1    Pinhasov, A.2    Mandel, S.3
  • 86
    • 0034083868 scopus 로고    scopus 로고
    • Activity-dependent neurotrophic factor: Intranasal administration of femtomolar-Acting peptides improve performance in a water maze
    • Gozes I, Giladi E, Pinhasov A, et al. Activity-dependent neurotrophic factor: intranasal administration of femtomolar-Acting peptides improve performance in a water maze. J Pharmacol Exp Ther. 2000; 293 (3): 1091-1098
    • (2000) J Pharmacol Exp Ther , vol.293 , Issue.3 , pp. 1091-1098
    • Gozes, I.1    Giladi, E.2    Pinhasov, A.3
  • 87
    • 79958128165 scopus 로고    scopus 로고
    • NAP (davunetide) provides functional and structural neuroprotection
    • Gozes I. NAP (davunetide) provides functional and structural neuroprotection. Curr Pharm Des. 2011; 17 (10): 1040-1044
    • (2011) Curr Pharm des , vol.17 , Issue.10 , pp. 1040-1044
    • Gozes, I.1
  • 88
    • 84895076670 scopus 로고    scopus 로고
    • NAP alpha-Aminoisobutyric acid (IsoNAP)
    • Gozes I, Schirer Y, Idan-Feldman A, et al. NAP alpha-Aminoisobutyric acid (IsoNAP). J Mol Neurosci. 2014; 52 (1): 1-9
    • (2014) J Mol Neurosci , vol.52 , Issue.1 , pp. 1-9
    • Gozes, I.1    Schirer, Y.2    Idan-Feldman, A.3
  • 89
    • 84902545124 scopus 로고    scopus 로고
    • Davunetide in patients with progressive supranuclear palsy: A randomised, double-blind, placebo-controlled phase 2/3 trial
    • Boxer AL, Lang AE, Grossman M, et al. Davunetide in patients with progressive supranuclear palsy: a randomised, double-blind, placebo-controlled phase 2/3 trial. Lancet Neurol. 2014; 13: 676-685
    • (2014) Lancet Neurol , vol.13 , pp. 676-685
    • Boxer, A.L.1    Lang, A.E.2    Grossman, M.3
  • 90
    • 33749614555 scopus 로고    scopus 로고
    • Tauopathy-like abnormalities and neurologic deficits in mice immunized with neuronal tau protein
    • Rosenmann H, Grigoriadis N, Karussis D, et al. Tauopathy-like abnormalities and neurologic deficits in mice immunized with neuronal tau protein. Arch Neurol. 2006; 63 (10): 1459-1467
    • (2006) Arch Neurol , vol.63 , Issue.10 , pp. 1459-1467
    • Rosenmann, H.1    Grigoriadis, N.2    Karussis, D.3
  • 91
    • 78650065372 scopus 로고    scopus 로고
    • Immunotherapy targeting pathological tau prevents cognitive decline in a new tangle mouse model
    • Boutajangout A, Quartermain D, Sigurdsson EM. Immunotherapy targeting pathological tau prevents cognitive decline in a new tangle mouse model. J Neurosci. 2010; 30 (49): 16559-16566
    • (2010) J Neurosci , vol.30 , Issue.49 , pp. 16559-16566
    • Boutajangout, A.1    Quartermain, D.2    Sigurdsson, E.M.3
  • 92
    • 84908290432 scopus 로고    scopus 로고
    • First-in-man tau vaccine targeting structural determinants essential for pathological tau-Tau interaction reduces tau oligomerisation and neurofibrillary degeneration in an Alzheimers disease model
    • Kontsekova E, Zilka N, Kovacech B, et al. First-in-man tau vaccine targeting structural determinants essential for pathological tau-Tau interaction reduces tau oligomerisation and neurofibrillary degeneration in an Alzheimers disease model. Alzheimers Res Ther. 2014; 6 (4): 44
    • (2014) Alzheimers Res Ther , vol.6 , Issue.4 , pp. 44
    • Kontsekova, E.1    Zilka, N.2    Kovacech, B.3
  • 93
    • 84922479255 scopus 로고    scopus 로고
    • Efficacy and safety of a liposome-based vaccine against protein Tau, assessed in tau. P301L mice that model tauopathy
    • Theunis C, Crespo-Biel N, Gafner V, et al. Efficacy and safety of a liposome-based vaccine against protein Tau, assessed in tau. P301L mice that model tauopathy. PLoS One. 2013; 8 (8): e72301
    • (2013) PLoS One , vol.8 , Issue.8 , pp. e72301
    • Theunis, C.1    Crespo-Biel, N.2    Gafner, V.3
  • 94
    • 79960563632 scopus 로고    scopus 로고
    • Passive immunization targeting pathological phospho-Tau protein in a mouse model reduces functional decline and clears tau aggregates from the brain
    • Boutajangout A, Ingadottir J, Davies P, et al. Passive immunization targeting pathological phospho-Tau protein in a mouse model reduces functional decline and clears tau aggregates from the brain. J Neurochem. 2011; 118 (4): 658-667
    • (2011) J Neurochem , vol.118 , Issue.4 , pp. 658-667
    • Boutajangout, A.1    Ingadottir, J.2    Davies, P.3
  • 95
    • 84890282160 scopus 로고    scopus 로고
    • Antibody uptake into neurons occurs primarily via clathrin-dependent Fcγ receptor endocytosis and is a prerequisite for acute tau protein clearance
    • Congdon EE, Gu J, Sait HB, et al. Antibody uptake into neurons occurs primarily via clathrin-dependent Fcγ receptor endocytosis and is a prerequisite for acute tau protein clearance. J Biol Chem. 2013; 288 (49): 35452-35465
    • (2013) J Biol Chem , vol.288 , Issue.49 , pp. 35452-35465
    • Congdon, E.E.1    Gu, J.2    Sait, H.B.3
  • 96
    • 84887837879 scopus 로고    scopus 로고
    • Two novel Tau antibodies targeting the 396/404 region are primarily taken up by neurons and reduce Tau protein pathology
    • Gu J, Congdon EE, Sigurdsson EM. Two novel Tau antibodies targeting the 396/404 region are primarily taken up by neurons and reduce Tau protein pathology. J Biol Chem. 2013; 288 (46): 33081-33095
    • (2013) J Biol Chem , vol.288 , Issue.46 , pp. 33081-33095
    • Gu, J.1    Congdon, E.E.2    Sigurdsson, E.M.3
  • 97
    • 68149124522 scopus 로고    scopus 로고
    • Chaperone-like antibodies in neurodegenerative tauopathies: Implication for immunotherapy
    • Kontsekova E, Ivanovova N, Handzusova M, et al. Chaperone-like antibodies in neurodegenerative tauopathies: implication for immunotherapy. Cell Mol Neurobiol. 2009; 29 (6-7): 793-798
    • (2009) Cell Mol Neurobiol , vol.29 , Issue.67 , pp. 793-798
    • Kontsekova, E.1    Ivanovova, N.2    Handzusova, M.3
  • 98
    • 20044388897 scopus 로고    scopus 로고
    • Effects of different anti-Tau antibodies on tau fibrillogenesis: RTA-1 and RTA-2 counteract tau aggregation
    • Taniguchi T, Sumida M, Hiraoka S, et al. Effects of different anti-Tau antibodies on tau fibrillogenesis: RTA-1 and RTA-2 counteract tau aggregation. FEBS Lett. 2005; 579 (6): 1399-1404
    • (2005) FEBS Lett , vol.579 , Issue.6 , pp. 1399-1404
    • Taniguchi, T.1    Sumida, M.2    Hiraoka, S.3
  • 99
    • 54249107245 scopus 로고    scopus 로고
    • Chaperone-like antibodies targeting misfolded tau protein: New vistas in the immunotherapy of neurodegenerative foldopathies
    • Zilka N, Kontsekova E, Novak M. Chaperone-like antibodies targeting misfolded tau protein: new vistas in the immunotherapy of neurodegenerative foldopathies. J Alzheimers Dis. 2008; 15: 169-179
    • (2008) J Alzheimers Dis , vol.15 , pp. 169-179
    • Zilka, N.1    Kontsekova, E.2    Novak, M.3
  • 100
    • 84908268752 scopus 로고    scopus 로고
    • Identification of structural determinants on tau protein essential for its pathological function: Novel therapeutic target for tau immunotherapy in Alzheimers disease
    • Kontsekova E, Zilka N, Kovacech B, et al. Identification of structural determinants on tau protein essential for its pathological function: novel therapeutic target for tau immunotherapy in Alzheimers disease. Alzheimers Res Ther. 2014; 6 (4): 45
    • (2014) Alzheimers Res Ther , vol.6 , Issue.4 , pp. 45
    • Kontsekova, E.1    Zilka, N.2    Kovacech, B.3
  • 101
    • 0033850407 scopus 로고    scopus 로고
    • Tau protein isoforms, phosphorylation and role in neurodegenerative disorders
    • Buée L, Bussière T, Buée-Scherrer V, et al. Tau protein isoforms, phosphorylation and role in neurodegenerative disorders. Brain Res Brain Res Rev. 2000; 33 (1): 95-130
    • (2000) Brain Res Brain Res Rev , vol.33 , Issue.1 , pp. 95-130
    • Buée, L.1    Bussière, T.2    Buée-Scherrer, V.3
  • 102
    • 84908377705 scopus 로고    scopus 로고
    • Neuronal uptake of tau/pS422 antibody and reduced progression of tau pathology in a mouse model of Alzheimers disease
    • Collin L, Bohrmann B, Göpfert U, et al. Neuronal uptake of tau/pS422 antibody and reduced progression of tau pathology in a mouse model of Alzheimers disease. Brain. 2014; 137 (Pt 10): 2834-2846
    • (2014) Brain , vol.137 , pp. 2834-2846
    • Collin, L.1    Bohrmann, B.2    Göpfert, U.3
  • 103
    • 84922785301 scopus 로고    scopus 로고
    • Human secreted tau increases amyloid-beta production
    • Bright J, Hussain S, Dang V, et al. Human secreted tau increases amyloid-beta production. Neurobiol Aging. 2015; 36 (2): 693-709
    • (2015) Neurobiol Aging , vol.36 , Issue.2 , pp. 693-709
    • Bright, J.1    Hussain, S.2    Dang, V.3
  • 104
    • 84919673394 scopus 로고    scopus 로고
    • Structure and mechanism of action of tau aggregation inhibitors
    • Cisek K, Cooper GL, Huseby CJ, et al. Structure and mechanism of action of tau aggregation inhibitors. Curr Alzheimer Res. 2014; 11 (10): 918-927
    • (2014) Curr Alzheimer Res , vol.11 , Issue.10 , pp. 918-927
    • Cisek, K.1    Cooper, G.L.2    Huseby, C.J.3
  • 105
    • 14844303721 scopus 로고    scopus 로고
    • Inhibition of heparin-induced Tau filament formation by phenothiazines, polyphenols, and porphyrins
    • Taniguchi S, Suzuki N, Masuda M, et al. Inhibition of heparin-induced Tau filament formation by phenothiazines, polyphenols, and porphyrins. J Biol Chem. 2005; 280 (9): 7614-7623
    • (2005) J Biol Chem , vol.280 , Issue.9 , pp. 7614-7623
    • Taniguchi, S.1    Suzuki, N.2    Masuda, M.3
  • 106
    • 84918784359 scopus 로고    scopus 로고
    • The green tea polyphenol (-)-epigallocatechin gallate prevents the aggregation of tau protein into toxic oligomers at substoichiometric ratios
    • Wobst HJ, Sharma A, Diamond MI, et al. The green tea polyphenol (-)-epigallocatechin gallate prevents the aggregation of tau protein into toxic oligomers at substoichiometric ratios. FEBS Lett. 2015; 589 (1): 77-83
    • (2015) FEBS Lett , vol.589 , Issue.1 , pp. 77-83
    • Wobst, H.J.1    Sharma, A.2    Diamond, M.I.3
  • 107
    • 34248142548 scopus 로고    scopus 로고
    • High throughput screening for small molecule inhibitors of heparin-induced tau fibril formation
    • Crowe A, Ballatore C, Hyde E, et al. High throughput screening for small molecule inhibitors of heparin-induced tau fibril formation. Biochem Biophys Res Commun. 2007; 358 (1): 1-6
    • (2007) Biochem Biophys Res Commun , vol.358 , Issue.1 , pp. 1-6
    • Crowe, A.1    Ballatore, C.2    Hyde, E.3
  • 108
    • 0029742937 scopus 로고    scopus 로고
    • Selective inhibition of Alzheimer disease-like tau aggregation by phenothiazines
    • Wischik CM, Edwards PC, Lai RY, et al. Selective inhibition of Alzheimer disease-like tau aggregation by phenothiazines. Proc Natl Acad Sci U S A. 1996; 93 (20): 11213-11218
    • (1996) Proc Natl Acad Sci U S A , vol.93 , Issue.20 , pp. 11213-11218
    • Wischik, C.M.1    Edwards, P.C.2    Lai, R.Y.3
  • 109
    • 1542327644 scopus 로고    scopus 로고
    • Ligand-dependent inhibition and reversal of Tau filament formation
    • Chirita C, Necula M, Kuret J. Ligand-dependent inhibition and reversal of Tau filament formation. Biochemistry. 2004; 43 (10): 2879-2887
    • (2004) Biochemistry , vol.43 , Issue.10 , pp. 2879-2887
    • Chirita, C.1    Necula, M.2    Kuret, J.3
  • 110
    • 23044439880 scopus 로고    scopus 로고
    • Cyanine dye N744 inhibits tau fibrillization by blocking filament extension: Implications for the treatment of tauopathic neurodegenerative diseases
    • Necula M, Chirita CN, Kuret J. Cyanine dye N744 inhibits tau fibrillization by blocking filament extension: implications for the treatment of tauopathic neurodegenerative diseases. Biochemistry. 2005; 44 (30): 10227-10237
    • (2005) Biochemistry , vol.44 , Issue.30 , pp. 10227-10237
    • Necula, M.1    Chirita, C.N.2    Kuret, J.3
  • 111
    • 66749176089 scopus 로고    scopus 로고
    • Structure-Activity relationship of cyanine tau aggregation inhibitors
    • Chang E, Congdon EE, Honson NS, et al. Structure-Activity relationship of cyanine tau aggregation inhibitors. J Med Chem. 2009; 52 (11): 3539-3547
    • (2009) J Med Chem , vol.52 , Issue.11 , pp. 3539-3547
    • Chang, E.1    Congdon, E.E.2    Honson, N.S.3
  • 112
    • 34548691220 scopus 로고    scopus 로고
    • N-phenylamine derivatives as aggregation inhibitors in cell models of tauopathy
    • Pickhardt M, Biernat J, Khlistunova I, et al. N-phenylamine derivatives as aggregation inhibitors in cell models of tauopathy. Curr Alzheimer Res. 2007; 4 (4): 397-402
    • (2007) Curr Alzheimer Res , vol.4 , Issue.4 , pp. 397-402
    • Pickhardt, M.1    Biernat, J.2    Khlistunova, I.3
  • 113
    • 77955927938 scopus 로고    scopus 로고
    • Tau protein and tau aggregation inhibitors
    • Bulic B, Pickhardt M, Mandelkow E-M, et al. Tau protein and tau aggregation inhibitors. Neuropharmacology. 2010; 59 (4-5): 276-289
    • (2010) Neuropharmacology , vol.59 , Issue.45 , pp. 276-289
    • Bulic, B.1    Pickhardt, M.2    Mandelkow, E.-M.3
  • 114
    • 37349128195 scopus 로고    scopus 로고
    • Rhodanine-based tau aggregation inhibitors in cell models of tauopathy
    • Bulic B, Pickhardt M, Khlistunova I, et al. Rhodanine-based tau aggregation inhibitors in cell models of tauopathy. Angew Chem Int Ed Engl. 2007; 46 (48): 9215-9219
    • (2007) Angew Chem Int Ed Engl , vol.46 , Issue.48 , pp. 9215-9219
    • Bulic, B.1    Pickhardt, M.2    Khlistunova, I.3
  • 115
    • 84887440374 scopus 로고    scopus 로고
    • Structural determinants of Tau aggregation inhibitor potency
    • Schafer KN, Cisek K, Huseby CJ, et al. Structural determinants of Tau aggregation inhibitor potency. J Biol Chem. 2013; 288 (45): 32599-32611
    • (2013) J Biol Chem , vol.288 , Issue.45 , pp. 32599-32611
    • Schafer, K.N.1    Cisek, K.2    Huseby, C.J.3
  • 116
    • 34347378629 scopus 로고    scopus 로고
    • Screening for inhibitors of tau protein aggregation into Alzheimer paired helical filaments: A ligand based approach results in successful scaffold hopping
    • Larbig G, Pickhardt M, Lloyd DG, et al. Screening for inhibitors of tau protein aggregation into Alzheimer paired helical filaments: a ligand based approach results in successful scaffold hopping. Curr Alzheimer Res. 2007; 4 (3): 315-323
    • (2007) Curr Alzheimer Res , vol.4 , Issue.3 , pp. 315-323
    • Larbig, G.1    Pickhardt, M.2    Lloyd, D.G.3
  • 117
    • 34548502687 scopus 로고    scopus 로고
    • Phenylthiazolyl-hydrazide and its derivatives are potent inhibitors of tau aggregation and toxicity in vitro and in cells
    • Pickhardt M, Larbig G, Khlistunova I, et al. Phenylthiazolyl-hydrazide and its derivatives are potent inhibitors of tau aggregation and toxicity in vitro and in cells. Biochemistry. 2007; 46: 10016-10023
    • (2007) Biochemistry , vol.46 , pp. 10016-10023
    • Pickhardt, M.1    Larbig, G.2    Khlistunova, I.3
  • 118
    • 13544251748 scopus 로고    scopus 로고
    • Anthraquinones inhibit tau aggregation and dissolve Alzheimers paired helical filaments in vitro and in cells
    • Pickhardt M, Gazova Z, von Bergen M, et al. Anthraquinones inhibit tau aggregation and dissolve Alzheimers paired helical filaments in vitro and in cells. J Biol Chem. 2005; 280 (5): 3628-3635
    • (2005) J Biol Chem , vol.280 , Issue.5 , pp. 3628-3635
    • Pickhardt, M.1    Gazova, Z.2    Von Bergen, M.3
  • 119
    • 68849086671 scopus 로고    scopus 로고
    • Identification of aminothienopyridazine inhibitors of tau assembly by quantitative high-Throughput screening
    • Crowe A, Huang W, Ballatore C, et al. Identification of aminothienopyridazine inhibitors of tau assembly by quantitative high-Throughput screening. Biochemistry. 2009; 48 (32): 7732-7745
    • (2009) Biochemistry , vol.48 , Issue.32 , pp. 7732-7745
    • Crowe, A.1    Huang, W.2    Ballatore, C.3
  • 120
    • 84863202528 scopus 로고    scopus 로고
    • Aminothienopyridazine inhibitors of tau aggregation: Evaluation of structure-Activity relationship leads to selection of candidates with desirable in vivo properties
    • Ballatore C, Crowe A, Piscitelli F, et al. Aminothienopyridazine inhibitors of tau aggregation: evaluation of structure-Activity relationship leads to selection of candidates with desirable in vivo properties. Bioorg Med Chem. 2012; 20 (14): 4451-4461
    • (2012) Bioorg Med Chem , vol.20 , Issue.14 , pp. 4451-4461
    • Ballatore, C.1    Crowe, A.2    Piscitelli, F.3
  • 121
    • 67651160771 scopus 로고    scopus 로고
    • Inhibition of tau fibrillization by oleocanthal via reaction with the amino groups of tau
    • Li W, Sperry JB, Crowe A, et al. Inhibition of tau fibrillization by oleocanthal via reaction with the amino groups of tau. J Neurochem. 2009; 110 (4): 1339-1351
    • (2009) J Neurochem , vol.110 , Issue.4 , pp. 1339-1351
    • Li, W.1    Sperry, J.B.2    Crowe, A.3
  • 122
    • 84867153806 scopus 로고    scopus 로고
    • Modulation of tau protein fibrillization by oleocanthal
    • Monti MC, Margarucci L, Riccio R, et al. Modulation of tau protein fibrillization by oleocanthal. J Nat Prod. 2012; 75 (9): 1584-1588
    • (2012) J Nat Prod , vol.75 , Issue.9 , pp. 1584-1588
    • Monti, M.C.1    Margarucci, L.2    Riccio, R.3
  • 123
    • 84925705424 scopus 로고    scopus 로고
    • Oleuropein aglycone: A possible drug against degenerative conditions. in vivo evidence of its effectiveness against Alzheimers disease
    • Casamenti F, Grossi C, Rigacci S, et al. Oleuropein aglycone: a possible drug against degenerative conditions. In vivo evidence of its effectiveness against Alzheimers disease. J Alzheimers Dis. 2015; 45 (3): 679-688
    • (2015) J Alzheimers Dis , vol.45 , Issue.3 , pp. 679-688
    • Casamenti, F.1    Grossi, C.2    Rigacci, S.3
  • 124
    • 84878869524 scopus 로고    scopus 로고
    • Interaction of cinnamaldehyde and epicatechin with tau: Implications of beneficial effects in modulating Alzheimers disease pathogenesis
    • George RC, Lew J, Graves DJ. Interaction of cinnamaldehyde and epicatechin with tau: implications of beneficial effects in modulating Alzheimers disease pathogenesis. J Alzheimers Dis. 2013; 36 (1): 21-40
    • (2013) J Alzheimers Dis , vol.36 , Issue.1 , pp. 21-40
    • George, R.C.1    Lew, J.2    Graves, D.J.3
  • 125
    • 67849127151 scopus 로고    scopus 로고
    • Cinnamon extract inhibits tau aggregation associated with Alzheimers disease in vitro
    • Peterson DW, George RC, Scaramozzino F, et al. Cinnamon extract inhibits tau aggregation associated with Alzheimers disease in vitro. J Alzheimers Dis. 2009; 17 (3): 585-597
    • (2009) J Alzheimers Dis , vol.17 , Issue.3 , pp. 585-597
    • Peterson, D.W.1    George, R.C.2    Scaramozzino, F.3
  • 126
    • 84892413120 scopus 로고    scopus 로고
    • Inhibition of Tau aggregation by three Aspergillus nidulans secondary metabolites: 2,ω-dihydroxyemodin, asperthecin, and asperbenzaldehyde
    • Paranjape SR, Chiang Y-M, Sanchez JF, et al. Inhibition of Tau aggregation by three Aspergillus nidulans secondary metabolites: 2,ω-dihydroxyemodin, asperthecin, and asperbenzaldehyde. Planta Med. 2014; 80 (1): 77-85
    • (2014) Planta Med , vol.80 , Issue.1 , pp. 77-85
    • Paranjape, S.R.1    Chiang, Y.-M.2    Sanchez, J.F.3
  • 127
    • 84865749287 scopus 로고    scopus 로고
    • Natural products as a rich source of tau-Targeting drugs for Alzheimers disease
    • Calcul L, Zhang B, Jinwal UK, et al. Natural products as a rich source of tau-Targeting drugs for Alzheimers disease. Future Med Chem. 2012; 4 (13): 1751-1761
    • (2012) Future Med Chem , vol.4 , Issue.13 , pp. 1751-1761
    • Calcul, L.1    Zhang, B.2    Jinwal, U.K.3
  • 128
    • 84880093048 scopus 로고    scopus 로고
    • Azaphilones: Chemistry and biology
    • Gao J-M, Yang S-X, Qin J-C. Azaphilones: chemistry and biology. Chem Rev. 2013; 113 (7): 4755-4811
    • (2013) Chem Rev , vol.113 , Issue.7 , pp. 4755-4811
    • Gao, J.-M.1    Yang, S.-X.2    Qin, J.-C.3
  • 129
    • 84930209014 scopus 로고    scopus 로고
    • Azaphilones inhibit tau aggregation and dissolve tau aggregates in vitro
    • Paranjape SR, Riley AP, Somoza AD, et al. Azaphilones inhibit tau aggregation and dissolve tau aggregates in vitro. ACS Chem Neurosci. 2015; 6 (5): 751-760
    • (2015) ACS Chem Neurosci , vol.6 , Issue.5 , pp. 751-760
    • Paranjape, S.R.1    Riley, A.P.2    Somoza, A.D.3
  • 130
    • 84876588286 scopus 로고    scopus 로고
    • Aminothienopyridazines and methylene blue affect Tau fibrillization via cysteine oxidation
    • Crowe A, James MJ, Lee VM, et al. Aminothienopyridazines and methylene blue affect Tau fibrillization via cysteine oxidation. J Biol Chem. 2013; 288 (16): 11024-11037
    • (2013) J Biol Chem , vol.288 , Issue.16 , pp. 11024-11037
    • Crowe, A.1    James, M.J.2    Lee, V.M.3
  • 131
    • 84936747253 scopus 로고    scopus 로고
    • The glutathione system: A new drug target in neuroimmune disorders
    • Morris G, Anderson G, Dean O, et al. The glutathione system: a new drug target in neuroimmune disorders. Mol Neurobiol. 2014; 50 (3): 1059-1084
    • (2014) Mol Neurobiol , vol.50 , Issue.3 , pp. 1059-1084
    • Morris, G.1    Anderson, G.2    Dean, O.3
  • 132
    • 70249102095 scopus 로고    scopus 로고
    • Modulation and detection of tau aggregation with small-molecule ligands
    • Oct;-414
    • Chang E, Honson NS, Bandyopadhyay B, et al. Modulation and detection of tau aggregation with small-molecule ligands. Curr Alzheimer Res. 2009 Oct; 6 (5): 409-414
    • (2009) Curr Alzheimer Res , vol.6 , Issue.5 , pp. 409
    • Chang, E.1    Honson, N.S.2    Bandyopadhyay, B.3
  • 133
    • 84879040199 scopus 로고    scopus 로고
    • Progress and developments in tau aggregation inhibitors for Alzheimer disease
    • Bulic B, Pickhardt M, Mandelkow E. Progress and developments in tau aggregation inhibitors for Alzheimer disease. J Med Chem. 2013; 56 (11): 4135-4155
    • (2013) J Med Chem , vol.56 , Issue.11 , pp. 4135-4155
    • Bulic, B.1    Pickhardt, M.2    Mandelkow, E.3
  • 134
    • 84863116610 scopus 로고    scopus 로고
    • Aggregation of α-synuclein is kinetically controlled by intramolecular diffusion
    • Ahmad B, Chen Y, Lapidus LJ. Aggregation of α-synuclein is kinetically controlled by intramolecular diffusion. Proc Natl Acad Sci U S A. 2012; 109 (7): 2336-2341
    • (2012) Proc Natl Acad Sci U S A , vol.109 , Issue.7 , pp. 2336-2341
    • Ahmad, B.1    Chen, Y.2    Lapidus, L.J.3
  • 135
    • 84858595914 scopus 로고    scopus 로고
    • Curcumin prevents aggregation in α-synuclein by increasing reconfiguration rate
    • Ahmad B, Lapidus LJ. Curcumin prevents aggregation in α-synuclein by increasing reconfiguration rate. J Biol Chem. 2012; 287 (12): 9193-9199
    • (2012) J Biol Chem , vol.287 , Issue.12 , pp. 9193-9199
    • Ahmad, B.1    Lapidus, L.J.2
  • 136
    • 84555177564 scopus 로고    scopus 로고
    • Clicked" sugar-curcumin conjugate: Modulator of amyloid-β and tau peptide aggregation at ultralow concentrations
    • Dolai S, Shi W, Corbo C, et al. "Clicked" sugar-curcumin conjugate: modulator of amyloid-β and tau peptide aggregation at ultralow concentrations. ACS Chem Neurosci. 2011; 2 (12): 694-699
    • (2011) ACS Chem Neurosci , vol.2 , Issue.12 , pp. 694-699
    • Dolai, S.1    Shi, W.2    Corbo, C.3
  • 137
    • 80055020556 scopus 로고    scopus 로고
    • Lysine-specific molecular tweezers are broad-spectrum inhibitors of assembly and toxicity of amyloid proteins
    • Sinha S, Lopes DH, Du Z, et al. Lysine-specific molecular tweezers are broad-spectrum inhibitors of assembly and toxicity of amyloid proteins. J Am Chem Soc. 2011; 133 (42): 16958-16969
    • (2011) J Am Chem Soc , vol.133 , Issue.42 , pp. 16958-16969
    • Sinha, S.1    Lopes, D.H.2    Du, Z.3
  • 138
    • 84871733093 scopus 로고    scopus 로고
    • Protection of primary neurons and mouse brain from Alzheimers pathology by molecular tweezers
    • Attar A, Ripoli C, Riccardi E, et al. Protection of primary neurons and mouse brain from Alzheimers pathology by molecular tweezers. Brain. 2012; 135 (12): 3735-3748
    • (2012) Brain , vol.135 , Issue.12 , pp. 3735-3748
    • Attar, A.1    Ripoli, C.2    Riccardi, E.3
  • 139
    • 84898619520 scopus 로고    scopus 로고
    • Molecular basis for preventing α-synuclein aggregation by a molecular tweezer
    • Acharya S, Safaie BM, Wongkongkathep P, et al. Molecular basis for preventing α-synuclein aggregation by a molecular tweezer. J Biol Chem. 2014; 289 (15): 10727-10737
    • (2014) J Biol Chem , vol.289 , Issue.15 , pp. 10727-10737
    • Acharya, S.1    Safaie, B.M.2    Wongkongkathep, P.3
  • 140
    • 79959791904 scopus 로고    scopus 로고
    • Towards a pharmacophore for amyloid
    • Landau M, Sawaya MR, Faull KF, et al. Towards a pharmacophore for amyloid. PLoS Biol. 2011; 9 (6): e1001080
    • (2011) PLoS Biol , vol.9 , Issue.6 , pp. e1001080
    • Landau, M.1    Sawaya, M.R.2    Faull, K.F.3
  • 141
    • 84874027197 scopus 로고    scopus 로고
    • Inhibition of tau filament formation by conformational modulation
    • Akoury E, Gajda M, Pickhardt M, et al. Inhibition of tau filament formation by conformational modulation. J Am Chem Soc. 2013; 135 (7): 2853-2862
    • (2013) J Am Chem Soc , vol.135 , Issue.7 , pp. 2853-2862
    • Akoury, E.1    Gajda, M.2    Pickhardt, M.3
  • 142
    • 33646570598 scopus 로고    scopus 로고
    • Small molecule inhibitors of alpha-synuclein filament assembly
    • Masuda M, Suzuki N, Taniguchi S, et al. Small molecule inhibitors of alpha-synuclein filament assembly. Biochemistry. 2006; 45 (19): 6085-6094
    • (2006) Biochemistry , vol.45 , Issue.19 , pp. 6085-6094
    • Masuda, M.1    Suzuki, N.2    Taniguchi, S.3
  • 143
    • 0001553682 scopus 로고
    • Color and constitution: One hundred years of research
    • Dähne S. Color and constitution: one hundred years of research. Science. 1978; 199 (4334): 1163-1167
    • (1978) Science , vol.199 , Issue.4334 , pp. 1163-1167
    • Dähne, S.1
  • 144
    • 79955674414 scopus 로고    scopus 로고
    • Lest we forget you-methylene blue
    • Schirmer RH, Adler H, Pickhardt M, et al. Lest we forget you-methylene blue. Neurobiol Aging. 2011; 32 (12): 2325.e7-2325.e16
    • (2011) Neurobiol Aging , vol.32 , Issue.12 , pp. 2325e7-2325e16
    • Schirmer, R.H.1    Adler, H.2    Pickhardt, M.3
  • 145
    • 20444413408 scopus 로고    scopus 로고
    • The phenothiazinium chromophore and the evolution of antimalarial drugs
    • Wainwright M, Amaral L. The phenothiazinium chromophore and the evolution of antimalarial drugs. Trop Med Int Health. 2005; 10 (6): 501-511
    • (2005) Trop Med Int Health , vol.10 , Issue.6 , pp. 501-511
    • Wainwright, M.1    Amaral, L.2
  • 146
    • 12344304237 scopus 로고    scopus 로고
    • Determination of methylene blue and leucomethylene blue in male and female Fischer 344 rat urine and B6C3F1 mouse urine
    • Gaudette NF, Lodge JW. Determination of methylene blue and leucomethylene blue in male and female Fischer 344 rat urine and B6C3F1 mouse urine. J Anal Toxicol. 2005; 29 (1): 28-33
    • (2005) J Anal Toxicol , vol.29 , Issue.1 , pp. 28-33
    • Gaudette, N.F.1    Lodge, J.W.2
  • 147
    • 67649817177 scopus 로고    scopus 로고
    • Strong gametocytocidal effect of methylene blue-based combination therapy against falciparum malaria: A randomised controlled trial
    • Coulibaly B, Zoungrana A, Mockenhaupt FP, et al. Strong gametocytocidal effect of methylene blue-based combination therapy against falciparum malaria: a randomised controlled trial. PLoS One. 2009; 4 (5): e5318
    • (2009) PLoS One , vol.4 , Issue.5 , pp. e5318
    • Coulibaly, B.1    Zoungrana, A.2    Mockenhaupt, F.P.3
  • 148
    • 77957016764 scopus 로고    scopus 로고
    • Role of monoamine oxidase, nitric oxide synthase and regional brain monoamines in the antidepressant-like effects of methylene blue and selected structural analogues
    • Harvey BH, Duvenhage I, Viljoen F, et al. Role of monoamine oxidase, nitric oxide synthase and regional brain monoamines in the antidepressant-like effects of methylene blue and selected structural analogues. Biochem Pharmacol. 2010; 80 (10): 1580-1591
    • (2010) Biochem Pharmacol , vol.80 , Issue.10 , pp. 1580-1591
    • Harvey, B.H.1    Duvenhage, I.2    Viljoen, F.3
  • 149
    • 0033948654 scopus 로고    scopus 로고
    • Pharmacokinetics and organ distribution of intravenous and oral methylene blue
    • Peter C, Hongwan D, Küpfer A, et al. Pharmacokinetics and organ distribution of intravenous and oral methylene blue. Eur J Clin Pharmacol. 2000; 56 (3): 247-250
    • (2000) Eur J Clin Pharmacol , vol.56 , Issue.3 , pp. 247-250
    • Peter, C.1    Hongwan, D.2    Küpfer, A.3
  • 150
    • 0031751618 scopus 로고    scopus 로고
    • Methylene blue supravital staining: An evaluation of its applicability to the mammalian brain and pineal gland
    • Müller T. Methylene blue supravital staining: an evaluation of its applicability to the mammalian brain and pineal gland. Histopathology. 1998; 13 (4): 1019-1026
    • (1998) Histopathology , vol.13 , Issue.4 , pp. 1019-1026
    • Müller, T.1
  • 151
    • 84919722777 scopus 로고    scopus 로고
    • Complex disposition of methylthioninium redox forms determines efficacy in tau aggregation inhibitor therapy for Alzheimers disease
    • Baddeley TC, McCaffrey J, Storey JM, et al. Complex disposition of methylthioninium redox forms determines efficacy in tau aggregation inhibitor therapy for Alzheimers disease. J Pharmacol Exp Ther. 2015; 352 (1): 110-118
    • (2015) J Pharmacol Exp Ther , vol.352 , Issue.1 , pp. 110-118
    • Baddeley, T.C.1    McCaffrey, J.2    Storey, J.M.3
  • 152
    • 84928401341 scopus 로고    scopus 로고
    • Cellular models of aggregation-dependent template-directed proteolysis to characterize Tau aggregation inhibitors for treatment of Alzheimer disease
    • Harrington CR, Storey JM, Clunas S, et al. Cellular models of aggregation-dependent template-directed proteolysis to characterize Tau aggregation inhibitors for treatment of Alzheimer disease. J Biol Chem. 2015; 290 (17): 10862-10875
    • (2015) J Biol Chem , vol.290 , Issue.17 , pp. 10862-10875
    • Harrington, C.R.1    Storey, J.M.2    Clunas, S.3
  • 153
    • 84928393250 scopus 로고    scopus 로고
    • Effects of oxidized and reduced forms of methylthioninium in two transgenic mouse tauopathy models
    • Melis V, Magbagbeolu M, Rickard JE, et al. Effects of oxidized and reduced forms of methylthioninium in two transgenic mouse tauopathy models. Behav Pharmacol. 2015; 26 (4): 353-368
    • (2015) Behav Pharmacol , vol.26 , Issue.4 , pp. 353-368
    • Melis, V.1    Magbagbeolu, M.2    Rickard, J.E.3
  • 154
    • 34248222543 scopus 로고    scopus 로고
    • Pharmacologic reductions of total tau levels; Implications for the role of microtubule dynamics in regulating tau expression
    • Dickey C, Ash P, Klosak N, et al. Pharmacologic reductions of total tau levels; implications for the role of microtubule dynamics in regulating tau expression. Mol Neurodegeneration. 2006; 1: 6
    • (2006) Mol Neurodegeneration , vol.1 , pp. 6
    • Dickey, C.1    Ash, P.2    Klosak, N.3
  • 155
    • 70349617699 scopus 로고    scopus 로고
    • Chemical manipulation of Hsp70 ATPase activity regulates tau stability
    • Jinwal UK, Miyata Y, Koren J 3rd, et al. Chemical manipulation of Hsp70 ATPase activity regulates tau stability. J Neurosci. 2009; 29 (39): 12079-12088
    • (2009) J Neurosci , vol.29 , Issue.39 , pp. 12079-12088
    • Jinwal, U.K.1    Miyata, Y.2    Koren, J.3
  • 156
    • 77958566761 scopus 로고    scopus 로고
    • Phenothiazine-mediated rescue of cognition in tau transgenic mice requires neuroprotection and reduced soluble tau burden
    • OLeary J, Li Q, Marinec P, et al. Phenothiazine-mediated rescue of cognition in tau transgenic mice requires neuroprotection and reduced soluble tau burden. Mol Neurodegener. 2010; 5: 45
    • (2010) Mol Neurodegener , vol.5 , pp. 45
    • Oleary, J.1    Li, Q.2    Marinec, P.3
  • 157
    • 84862281225 scopus 로고    scopus 로고
    • Methylthioninium chloride (methylene blue) induces autophagy and attenuates tauopathy in vitro and in vivo
    • Congdon EE, Wu JW, Myeku N, et al. Methylthioninium chloride (methylene blue) induces autophagy and attenuates tauopathy in vitro and in vivo. Autophagy. 2012; 8 (4): 609-622
    • (2012) Autophagy , vol.8 , Issue.4 , pp. 609-622
    • Congdon, E.E.1    Wu, J.W.2    Myeku, N.3
  • 158
    • 84876433114 scopus 로고    scopus 로고
    • Methylene blue induces macroautophagy through 5 adenosine monophosphate-Activated protein kinase pathway to protect neurons from serum deprivation
    • Xie L, Li W, Winters A, et al. Methylene blue induces macroautophagy through 5 adenosine monophosphate-Activated protein kinase pathway to protect neurons from serum deprivation. Front Cell Neurosci. 2013; 7: 56
    • (2013) Front Cell Neurosci , vol.7 , pp. 56
    • Xie, L.1    Li, W.2    Winters, A.3
  • 159
    • 84875182760 scopus 로고    scopus 로고
    • Mechanistic basis of phenothiazine-driven inhibition of tau aggregation
    • Akoury E, Pickhardt M, Gajda M, et al. Mechanistic basis of phenothiazine-driven inhibition of tau aggregation. Angewandte Chemie Int Ed. 2013; 52 (12): 3511-3515
    • (2013) Angewandte Chemie Int Ed , vol.52 , Issue.12 , pp. 3511-3515
    • Akoury, E.1    Pickhardt, M.2    Gajda, M.3
  • 160
    • 0030804625 scopus 로고    scopus 로고
    • The interaction between methylene blue and the cholinergic system
    • Pfaffendorf M, Bruning TA, Batink HD, et al. The interaction between methylene blue and the cholinergic system. Br J Pharmacol. 1997; 122 (1): 95-98
    • (1997) Br J Pharmacol , vol.122 , Issue.1 , pp. 95-98
    • Pfaffendorf, M.1    Bruning, T.A.2    Batink, H.D.3
  • 161
    • 0027533361 scopus 로고
    • Inhibition of nitric oxide synthesis by methylene blue
    • Mayer B, Brunner F, Schmidt K. Inhibition of nitric oxide synthesis by methylene blue. Biochem Pharmacol. 1993; 45 (2): 367-374
    • (1993) Biochem Pharmacol , vol.45 , Issue.2 , pp. 367-374
    • Mayer, B.1    Brunner, F.2    Schmidt, K.3
  • 162
    • 0141725416 scopus 로고    scopus 로고
    • Pharmacological evidence that methylene blue inhibits noradrenaline neuronal uptake in the rat vas deferens
    • Chies AB, Custódio RC, de Souza GL, et al. Pharmacological evidence that methylene blue inhibits noradrenaline neuronal uptake in the rat vas deferens. Pol J Pharmacol. 2003; 55 (4): 573-579
    • (2003) Pol J Pharmacol , vol.55 , Issue.4 , pp. 573-579
    • Chies, A.B.1    Custódio, R.C.2    De Souza, G.L.3
  • 163
    • 41149175103 scopus 로고    scopus 로고
    • Adverse effects of methylene blue on the central nervous system
    • Vutskits L, Briner A, Klauser P, et al. Adverse effects of methylene blue on the central nervous system. Anesthesiology. 2008; 108 (4): 684-692
    • (2008) Anesthesiology , vol.108 , Issue.4 , pp. 684-692
    • Vutskits, L.1    Briner, A.2    Klauser, P.3
  • 164
    • 36048982476 scopus 로고    scopus 로고
    • Methylene blue and serotonin toxicity: Inhibition of monoamine oxidase A (MAO A) confirms a theoretical prediction
    • Ramsay RR, Dunford C, Gillman PK. Methylene blue and serotonin toxicity: inhibition of monoamine oxidase A (MAO A) confirms a theoretical prediction. Br J Pharmacol. 2007; 152 (6): 946-951
    • (2007) Br J Pharmacol , vol.152 , Issue.6 , pp. 946-951
    • Ramsay, R.R.1    Dunford, C.2    Gillman, P.K.3
  • 165
    • 34547672881 scopus 로고    scopus 로고
    • Methylene blue inhibits amyloid Abeta oligomerization by promoting fibrillization
    • Necula M, Breydo L, Milton S, et al. Methylene blue inhibits amyloid Abeta oligomerization by promoting fibrillization. Biochemistry. 2007; 46 (30): 8850-8860
    • (2007) Biochemistry , vol.46 , Issue.30 , pp. 8850-8860
    • Necula, M.1    Breydo, L.2    Milton, S.3
  • 166
    • 79251555689 scopus 로고    scopus 로고
    • Methylene blue reduces aβ levels and rescues early cognitive deficit by increasing proteasome activity
    • Medina DX, Caccamo A, Oddo S. Methylene blue reduces aβ levels and rescues early cognitive deficit by increasing proteasome activity. Brain Pathol. 2011; 21 (2): 140-149
    • (2011) Brain Pathol , vol.21 , Issue.2 , pp. 140-149
    • Medina, D.X.1    Caccamo, A.2    Oddo, S.3
  • 167
    • 59449097789 scopus 로고    scopus 로고
    • Methylthioninium chloride reverses cognitive deficits induced by scopolamine: Comparison with rivastigmine
    • Deiana S, Harrington CR, Wischik CM, et al. Methylthioninium chloride reverses cognitive deficits induced by scopolamine: comparison with rivastigmine. Psychopharmacology (Berl). 2009; 202 (1-3): 53-65
    • (2009) Psychopharmacology (Berl) , vol.202 , Issue.13 , pp. 53-65
    • Deiana, S.1    Harrington, C.R.2    Wischik, C.M.3
  • 168
    • 78650939779 scopus 로고    scopus 로고
    • Beneficial network effects of methylene blue in an amnestic model
    • Riha PD, Rojas JC, Gonzalez-Lima F. Beneficial network effects of methylene blue in an amnestic model. Neuroimage. 2011; 54 (4): 2623-2634
    • (2011) Neuroimage , vol.54 , Issue.4 , pp. 2623-2634
    • Riha, P.D.1    Rojas, J.C.2    Gonzalez-Lima, F.3
  • 169
    • 84886597463 scopus 로고    scopus 로고
    • Therapeutic and preventive effects of methylene blue on Alzheimers disease pathology in a transgenic mouse model
    • PT A
    • Paban V, Manrique C, Filali M, et al. Therapeutic and preventive effects of methylene blue on Alzheimers disease pathology in a transgenic mouse model. Neuropharmacology. 2014; 76 (Pt A): 68-79
    • (2014) Neuropharmacology , vol.76 , pp. 68-79
    • Paban, V.1    Manrique, C.2    Filali, M.3
  • 170
    • 84910026958 scopus 로고    scopus 로고
    • Methylene blue modulates β-secretase, reverses cerebral amyloidosis, and improves cognition in transgenic mice
    • Mori T, Koyama N, Segawa T, et al. Methylene blue modulates β-secretase, reverses cerebral amyloidosis, and improves cognition in transgenic mice. J Biol Chem. 2014; 289 (44): 30303-30317
    • (2014) J Biol Chem , vol.289 , Issue.44 , pp. 30303-30317
    • Mori, T.1    Koyama, N.2    Segawa, T.3
  • 171
    • 84958123568 scopus 로고    scopus 로고
    • Methylene blue improves brain mitochondrial ABAD functions and decreases Aβ in a neuroinflammatory Alzheimers disease mouse model
    • [Epub ahead of print]. doi: 10.1007/s12035-014-9088-8
    • Zakaria A, Hamdi N, Abdel-Kader RM. Methylene blue improves brain mitochondrial ABAD functions and decreases Aβ in a neuroinflammatory Alzheimers disease mouse model. Mol Neurobiol. 2015. [Epub ahead of print]. doi: 10.1007/s12035-014-9088-8
    • (2015) Mol Neurobiol
    • Zakaria, A.1    Hamdi, N.2    Abdel-Kader, R.M.3
  • 172
    • 84927639610 scopus 로고    scopus 로고
    • Methylene blue protects astrocytes against glucose oxygen deprivation by improving cellular respiration
    • Roy Choudhury G, Winters A, Rich RM, et al. Methylene blue protects astrocytes against glucose oxygen deprivation by improving cellular respiration. PLoS One. 2015; 10 (4): e0123096
    • (2015) PLoS One , vol.10 , Issue.4 , pp. e0123096
    • Roy Choudhury, G.1    Winters, A.2    Rich, R.M.3
  • 173
    • 79955528923 scopus 로고    scopus 로고
    • Alternative mitochondrial electron transfer as a novel strategy for neuroprotection
    • Wen Y, Li W, Poteet EC, et al. Alternative mitochondrial electron transfer as a novel strategy for neuroprotection. J Biol Chem. 2011; 286 (18): 16504-16515
    • (2011) J Biol Chem , vol.286 , Issue.18 , pp. 16504-16515
    • Wen, Y.1    Li, W.2    Poteet, E.C.3
  • 174
    • 84868282410 scopus 로고    scopus 로고
    • Neuroprotective actions of methylene blue and its derivatives
    • Poteet E, Winters A, Yan LJ, et al. Neuroprotective actions of methylene blue and its derivatives. PLoS One. 2012; 7 (10): e48279
    • (2012) PLoS One , vol.7 , Issue.10 , pp. e48279
    • Poteet, E.1    Winters, A.2    Yan, L.J.3
  • 175
    • 84902953435 scopus 로고    scopus 로고
    • Methylene blue upregulates Nrf2/ARE genes and prevents tau-related neurotoxicity
    • Stack C, Jainuddin S, Elipenahli C, et al. Methylene blue upregulates Nrf2/ARE genes and prevents tau-related neurotoxicity. Hum Mol Genet. 2014; 23 (14): 3716-3732
    • (2014) Hum Mol Genet , vol.23 , Issue.14 , pp. 3716-3732
    • Stack, C.1    Jainuddin, S.2    Elipenahli, C.3
  • 176
    • 85018215020 scopus 로고    scopus 로고
    • Preventive methylene blue treatment preserves cognition in mice expressing full-length pro-Aggregant human Tau
    • Hochgräfe K, Sydow A, Matenia D, et al. Preventive methylene blue treatment preserves cognition in mice expressing full-length pro-Aggregant human Tau. Acta Neuropathol Commun. 2015; 3: 25
    • (2015) Acta Neuropathol Commun , vol.3 , pp. 25
    • Hochgräfe, K.1    Sydow, A.2    Matenia, D.3
  • 177
    • 84934882160 scopus 로고    scopus 로고
    • Nontoxic singlet oxygen generator as a therapeutic candidate for treating tauopathies
    • Mohideen SS, Yamasaki Y, Omata Y, et al. Nontoxic singlet oxygen generator as a therapeutic candidate for treating tauopathies. Sci Rep. 2015; 5: 10821
    • (2015) Sci Rep , vol.5 , pp. 10821
    • Mohideen, S.S.1    Yamasaki, Y.2    Omata, Y.3
  • 178
    • 84868087867 scopus 로고    scopus 로고
    • Methylene blue inhibits the function of α7-nicotinic acetylcholine receptors
    • Al Mansouri AS, Lorke DE, Nurulain SM, et al. Methylene blue inhibits the function of α7-nicotinic acetylcholine receptors. CNS Neurol Disord Drug Targets. 2012; 11 (6): 791-800
    • (2012) CNS Neurol Disord Drug Targets , vol.11 , Issue.6 , pp. 791-800
    • Al Mansouri, A.S.1    Lorke, D.E.2    Nurulain, S.M.3
  • 179
    • 0030805455 scopus 로고    scopus 로고
    • Stimulation of respiration by methylene blue in rat liver mitochondria
    • Visarius TM, Stucki JW, Lauterburg BH. Stimulation of respiration by methylene blue in rat liver mitochondria. FEBS Lett. 1997; 412 (1): 157-160
    • (1997) FEBS Lett , vol.412 , Issue.1 , pp. 157-160
    • Visarius, T.M.1    Stucki, J.W.2    Lauterburg, B.H.3
  • 180
    • 77954954324 scopus 로고    scopus 로고
    • Methylene blue fails to inhibit Tau and polyglutamine protein dependent toxicity in zebrafish
    • Sep;-271
    • van Bebber F, Paquet D, Hruscha A, et al. Methylene blue fails to inhibit Tau and polyglutamine protein dependent toxicity in zebrafish. Neurobiol Dis. 2010 Sep; 39 (3): 265-271
    • (2010) Neurobiol Dis , vol.39 , Issue.3 , pp. 265
    • Van Bebber, F.1    Paquet, D.2    Hruscha, A.3
  • 181
    • 84871446624 scopus 로고    scopus 로고
    • Methylene blue reduced abnormal tau accumulation in P301L tau transgenic mice
    • Hosokawa M, Arai T, Masuda-Suzukake M, et al. Methylene blue reduced abnormal tau accumulation in P301L tau transgenic mice. PLoS One. 2012; 7 (12): e52389
    • (2012) PLoS One , vol.7 , Issue.12 , pp. e52389
    • Hosokawa, M.1    Arai, T.2    Masuda-Suzukake, M.3
  • 182
    • 84893473873 scopus 로고    scopus 로고
    • Methylene blue does not reverse existing neurofibrillary tangle pathology in the rTg4510 mouse model of tauopathy
    • Spires-Jones TL, Friedman T, Pitstick R, et al. Methylene blue does not reverse existing neurofibrillary tangle pathology in the rTg4510 mouse model of tauopathy. Neurosci Lett. 2014; 562: 63-68
    • (2014) Neurosci Lett , vol.562 , pp. 63-68
    • Spires-Jones, T.L.1    Friedman, T.2    Pitstick, R.3
  • 183
    • 84868537186 scopus 로고    scopus 로고
    • Binding of methylene blue to a surface cleft inhibits the oligomerization and fibrillization of prion protein
    • Cavaliere P, Torrent J, Prigent S, et al. Binding of methylene blue to a surface cleft inhibits the oligomerization and fibrillization of prion protein. Biochim Biophys Acta. 2013; 1832 (1): 20-28
    • (2013) Biochim Biophys Acta , vol.1832 , Issue.1 , pp. 20-28
    • Cavaliere, P.1    Torrent, J.2    Prigent, S.3
  • 184
    • 67650140610 scopus 로고    scopus 로고
    • Methylene blue and dimebon inhibit aggregation of TDP-43 in cellular models
    • Yamashita M, Nonaka T, Arai T, et al. Methylene blue and dimebon inhibit aggregation of TDP-43 in cellular models. FEBS Lett. 2009; 583 (14): 2419-2424
    • (2009) FEBS Lett , vol.583 , Issue.14 , pp. 2419-2424
    • Yamashita, M.1    Nonaka, T.2    Arai, T.3
  • 185
    • 77949908515 scopus 로고    scopus 로고
    • Phosphorylated and cleaved TDP-43 in ALS, FTLD and other neurodegenerative disorders and in cellular models of TDP-43 proteinopathy
    • Arai T, Hasegawa M, Nonoka T, et al. Phosphorylated and cleaved TDP-43 in ALS, FTLD and other neurodegenerative disorders and in cellular models of TDP-43 proteinopathy. Neuropathology. 2010; 30 (2): 170-181
    • (2010) Neuropathology , vol.30 , Issue.2 , pp. 170-181
    • Arai, T.1    Hasegawa, M.2    Nonoka, T.3
  • 186
    • 80855143691 scopus 로고    scopus 로고
    • Novel types of frontotemporal lobar degeneration: Beyond tau and TDP-43
    • Mackenzie IRA, Neumann M, Cairns NJ, et al. Novel types of frontotemporal lobar degeneration: beyond tau and TDP-43. J Mol Neurosci. 2011; 45 (3): 402-408
    • (2011) J Mol Neurosci , vol.45 , Issue.3 , pp. 402-408
    • Mackenzie, I.R.A.1    Neumann, M.2    Cairns, N.J.3
  • 187
    • 84864862868 scopus 로고    scopus 로고
    • Methylene blue modulates huntingtin aggregation intermediates and is protective in Huntingtons disease models
    • Sontag EM, Lotz GP, Agrawal N, et al. Methylene blue modulates huntingtin aggregation intermediates and is protective in Huntingtons disease models. J Neurosci. 2012; 32 (32): 11109-11119
    • (2012) J Neurosci , vol.32 , Issue.32 , pp. 11109-11119
    • Sontag, E.M.1    Lotz, G.P.2    Agrawal, N.3
  • 188
    • 84921531217 scopus 로고    scopus 로고
    • Tau aggregation inhibitor therapy: An exploratory phase 2 study in mild or moderate Alzheimers disease
    • Wischik CM, Staff RT, Wischik DJ, et al. Tau aggregation inhibitor therapy: an exploratory phase 2 study in mild or moderate Alzheimers disease. J Alzheimers Dis. 2015; 44 (2): 705-720
    • (2015) J Alzheimers Dis , vol.44 , Issue.2 , pp. 705-720
    • Wischik, C.M.1    Staff, R.T.2    Wischik, D.J.3
  • 189
    • 84861372942 scopus 로고    scopus 로고
    • The crystalline state of methylene blue: A zoo of hydrates
    • Rager T, Geoffroy A, Hilfiker R, et al. The crystalline state of methylene blue: a zoo of hydrates. Phys Chem Chem Phys. 2012; 14 (22): 8074-8082
    • (2012) Phys Chem Chem Phys , vol.14 , Issue.22 , pp. 8074-8082
    • Rager, T.1    Geoffroy, A.2    Hilfiker, R.3
  • 190
    • 84928386659 scopus 로고    scopus 로고
    • Different pathways of molecular pathophysiology underlie cognitive and motor tauopathy phenotypes in transgenic models for Alzheimers disease and frontotemporal lobar degeneration
    • Melis V, Zabke C, Stamer K, et al. Different pathways of molecular pathophysiology underlie cognitive and motor tauopathy phenotypes in transgenic models for Alzheimers disease and frontotemporal lobar degeneration. Cell Mol Life Sci. 2015; 72 (11): 2199-2222
    • (2015) Cell Mol Life Sci , vol.72 , Issue.11 , pp. 2199-2222
    • Melis, V.1    Zabke, C.2    Stamer, K.3
  • 191
    • 33846212717 scopus 로고    scopus 로고
    • Kinases and phosphatases and tau sites involved in Alzheimer neurofibrillary degeneration
    • Wang JZ, Grundke-Iqbal I, Iqbal K. Kinases and phosphatases and tau sites involved in Alzheimer neurofibrillary degeneration. Eur J Neurochem. 2007; 25 (1): 59-68
    • (2007) Eur J Neurochem , vol.25 , Issue.1 , pp. 59-68
    • Wang, J.Z.1    Grundke-Iqbal, I.2    Iqbal, K.3
  • 192
    • 84890392083 scopus 로고    scopus 로고
    • New insights into the role of glycogen synthase kinase-3 in Alzheimers disease
    • Medina M, Avila J. New insights into the role of glycogen synthase kinase-3 in Alzheimers disease. Expert Opin Ther Targets. 2014; 18 (1): 69-77
    • (2014) Expert Opin Ther Targets , vol.18 , Issue.1 , pp. 69-77
    • Medina, M.1    Avila, J.2
  • 193
    • 84943637753 scopus 로고    scopus 로고
    • Critical role of acetylation in tau-mediated neurodegeneration and cognitive deficits
    • Min S-W, Chen X, Tracy TE, et al. Critical role of acetylation in tau-mediated neurodegeneration and cognitive deficits. Nat Med. 2015; 21: 1154-1162. doi: 10.1038/nm.3951
    • (2015) Nat Med , vol.21 , pp. 1154-1162
    • Min, S.-W.1    Chen, X.2    Tracy, T.E.3
  • 194
    • 84896837399 scopus 로고    scopus 로고
    • Tau immunotherapy and imaging
    • Sigurdsson EM. Tau immunotherapy and imaging. Neurodegener Dis. 2014; 13 (2-3): 103-106
    • (2014) Neurodegener Dis , vol.13 , Issue.23 , pp. 103-106
    • Sigurdsson, E.M.1
  • 195
    • 84896269359 scopus 로고    scopus 로고
    • Passive immunization with Tau oligomer monoclonal antibody reverses tauopathy phenotypes without affecting hyperphosphorylated neurofibrillary tangles
    • Castillo-Carranza DL, Sengupta U, Guerrero-Muñoz MJ, et al. Passive immunization with Tau oligomer monoclonal antibody reverses tauopathy phenotypes without affecting hyperphosphorylated neurofibrillary tangles. J Neurosci. 2014; 34 (12): 4260-4272
    • (2014) J Neurosci , vol.34 , Issue.12 , pp. 4260-4272
    • Castillo-Carranza, D.L.1    Sengupta, U.2    Guerrero-Muñoz, M.J.3
  • 196
    • 84941005081 scopus 로고    scopus 로고
    • Longitudinal cerebrospinal fluid biomarker changes in preclinical Alzheimer disease during middle age
    • Sutphen CL, Jasielec MS, Shah AR, et al. Longitudinal cerebrospinal fluid biomarker changes in preclinical Alzheimer disease during middle age. JAMA Neurol. 2015; 72 (9): 1029-1042
    • (2015) JAMA Neurol , vol.72 , Issue.9 , pp. 1029-1042
    • Sutphen, C.L.1    Jasielec, M.S.2    Shah, A.R.3
  • 197
    • 84884127276 scopus 로고    scopus 로고
    • Preclinical Alzheimers disease and its outcome: A longitudinal cohort study
    • Vos SJ, Xiong C, Visser PJ, et al. Preclinical Alzheimers disease and its outcome: a longitudinal cohort study. Lancet Neurol. 2013; 12 (10): 957-965
    • (2013) Lancet Neurol , vol.12 , Issue.10 , pp. 957-965
    • Vos, S.J.1    Xiong, C.2    Visser, P.J.3


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.